NZ508018A - DNA construct containing AD7c-NTP sequence controlled by neurospecific promoter to treat Alzheimer's disease and neural tumours - Google Patents

DNA construct containing AD7c-NTP sequence controlled by neurospecific promoter to treat Alzheimer's disease and neural tumours

Info

Publication number
NZ508018A
NZ508018A NZ508018A NZ50801800A NZ508018A NZ 508018 A NZ508018 A NZ 508018A NZ 508018 A NZ508018 A NZ 508018A NZ 50801800 A NZ50801800 A NZ 50801800A NZ 508018 A NZ508018 A NZ 508018A
Authority
NZ
New Zealand
Prior art keywords
ntp
ad7c
seq
cells
expression
Prior art date
Application number
NZ508018A
Inventor
La Monte Suzanne De
Jack R Wands
Original Assignee
Gen Hospital Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Gen Hospital Corp filed Critical Gen Hospital Corp
Publication of NZ508018A publication Critical patent/NZ508018A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0312Animal model for Alzheimer's disease
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0393Animal model comprising a reporter system for screening tests

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Neurology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Neurosurgery (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)

Abstract

Disclosed are transgenic animals and transfected cell lines expressing a protein associated with Alzheimer's disease, neuroectodermal tumours, malignant astrocytomas, and glioblastomas. Also disclosed is the use of such transgenic animals and transfected cell lines to screen potential drug candidates for treating or preventing Alzheimer's disease, neuroectodermal tumours, malignant astrocytomas, and glioblastomas. Further described are antisense oligonucleotides, ribozymes, triplex forming DNA and external guide sequences that can be used to treat or prevent Alzheimer's disease, neuroectodermal tumours, malignant astrocytomas, and glioblastomas in non-human animals, (62) Divided out of 337445

Description

<div class="application article clearfix" id="description"> <p class="printTableText" lang="en">0 1 <br><br> NEW ZEALAND <br><br> PATENTS ACT, 1953 <br><br> No: Date: <br><br> Divided out of NZ 337445 <br><br> Dated 26 February 1998 <br><br> COMPLETE SPECIFICATION <br><br> TRANSGENIC ANIMALS AND CELL LINES FOR SCREENING DRUGS EFFECTIVE FOR THE TREATMENT OR PREVENTION OF ALZHEIMER'S DISEASE <br><br> We, THE GENERAL HOSPITAL CORPORATION a corporation of the State of Massachusetts, United States of America, of Fruit Street, Boston, MA 02114, United States of America, do hereby declare the invention for which we pray that a patent may be granted to us, and the method by which it is to be performed, to be particularly described in and by the following statement: <br><br> 1 <br><br> (followed by page la) <br><br> INTELLECTUAL PROPERTY OFFICE OF N.Z. <br><br> - 8 NOV 2000 <br><br> Transgenic Animals and Cell Lines for Screening Drugs Effective for the Treatment or Prevention of Alzheimer's <br><br> Disease <br><br> Background of the Invention <br><br> Statement as to Rights to Inventions Made Under Federally-Sponsored Research and Development <br><br> This invention was made with U.S. Government support under grant nos. CA-35711, AA-00026 and AA-002169, awarded by the National Institutes of Health. The U.S. Government has certain rights in this invention. <br><br> Field of the Invention <br><br> The present invention is in the field of genetic engineering and molecular biology. In particular, the invention is directed to transgenic animals and transfected cell lines expressing a protein associated with Alzheimer's Disease, neuroectodermal tumors, malignant astrocytomas, and glioblastomas. This invention is also directed to the use of such transgenic animals and transfected cell lines to screen potential drug candidates for treating or preventing Alzheimer's disease. The invention also relates to new antisense oligonucleotides, ribozymes, triplex forming DNA and external guide sequences that can be used to treat or prevent Alzheimer's disease. <br><br> Related Art <br><br> Alzheimer's disease (AD) (Khachaturian, Z.S., "Diagnosis of Alzheimer's Disease," Arch. Neurol. 427:1097-1105 (1985)) is the most prevalent neurodegenerative disease and the most common cause of dementia in the W estern <br><br> hemisphere. AD neurodegeneration is characterized by prominent atrophy of corticolimbic structures with neuronal loss, neurofibrillary tangle formation, aberrant proliferation of neurites, senile plaques, and p A4-amyloid deposition in the brain (Khachaturian, Z.S.). Approximately 90 percent of AD occurs sporadically. The cause is unknown, but the most important overall risk factor is aging (Takman, A., "Epidemiology of Alzheimer's Disease. Issues of Etiology and Validity,n Acta Neurol. Scand. Suppl. 745:1-70(1993)). The apolipoprotein €4 genotype (Corder, E.H. et al., "Gene Does of Apolipoprotein E Type 4 Allele and the Risk of Alzheimer,' s Disease in Late Onset Families, Science 261:921 -923 (1993)) and a family history of Trisomy 21 Down syndrome (Lai, F. and Williams, R.S., "A Prospective Study of Alzheimer Disease in Down Syndrome," Arch. Neurol. 46:849-853 (1989) increase risk or accelerate the course of sporadic AD. Familial forms of AD, which account for 5 to 10 percent of the cases, have been linked to mutations in the amyloid precursor protein (APP) gene (Kennedy, A.M. etal, "Familial Alzheimer's Disease. A Pedigree With a Mis-Sense Mutation in the Amyloid Precursor Protein Gene (Amyloid Precursor Protein 717 Valine --&gt; Glycine," Brain 309-324 (1993); Peacock, M.L. elal., "Novel Amyloid Precursor Protein Gene Mutation (Codon 66 5 Asp) in a Patient with Late-Onset Alzheimer's Disease." Ann. Neurol. 35:432-438 (1994); Tanzi, R.E. ei al., "Assessment of Amyloid Beta-Protein Precursor Gene Mutations in a Large Set of Familial and Sporadic Alzheimer's Disease Cases,"Am. J. Hum. Genet. 51:273-282 (1992)) located on Chromosome 21 (Robakis, N.K. et al.. Chromosome 21q21 Sub localization of Gene Encoding Beta-Amyloid Peptide in Cerebral Vessels and Neuritic (Senile) Plaques of People with Alzheimer Disease and Down Syndrome," Lancet 1:384-385 (1987)), or presenilin genes located on Chromosomes 1 and 14 (Levy-Lahad, E. et al, "Candidate Gene for the Chromosome 1 Familial Alzheimer's Disease Locus," Science 75:973-977(1995); Sorbi, S. et al., "Missense Mutation of SI 82 Gene in Italian Families With Early Onset Alzheimer's Disease," Lancet 346:439-440 (1995); Sherrington, R. et al., "Cloning of a Gene Bearing Missense Mutations in Early-Onset Familial <br><br> Alzheimer's Disease, Nature 375:754-760 (1995); Rogaev, E.I. et al., "Familial Alzheimer's Disease in Kindreds With Missense Mutations in a Gene on Chromosome 1 Related to the Alzheimer's Disease Type 3 Gene," Nature 57(5:775-778 (1995); Barinaga, M. et al., "Candidate Gene for the Chromosome 1 Familial Alzheimer's Disease Locus," Science 269:973-977 (1995)). Over-expression and abnormal cleavage of APP may promote AD neurodegeneration since all individuals with Trisomy 21 Down syndrome who survive beyond the fourth decade develop AD with extensive central nervous system (CNS) accumulations of PA4-amyloid (Lai, F. and Williams, R.S., "A Prospective Study of Alzheimer Disease in Down Syndrome," Arch. Neurol. 46:849-853 (1989)), and experimentally, PA4-amyloid is neurotoxic and apotogenic (LaFerla, F.M. et al., "The Alzheimer's A Beta Peptide Induces Neurodegeneration and Apoptotic Cell Death in Transgenic Mice," Nat. Genet. 9:21-30 (1995). In addition, missense mutations in persenilin 1. as occurs in nature, cause vasculopathy and massive accumulations of peptides in the brain (Lemere, C.A. et al., "The E280A Presenilin 1 Alzheimer Mutation Produces Increased AP42 Deposition and Severe Cerebellar Pathology," Nature Med. 2:1146-1150 (1996); Mann. D.M. et al.. "Amyloid Beta Protein (Abeta) Deposition in Chromosome 14-Linked Alzheimer's Disease: Predominance of Abeta42(43)," Ann Neurol. 40:149-156 (1996)). <br><br> Central nervous system biochemical and molecular abnormalities identified in AD include: 1) increased phosphorylation of tow and other cytoskeletal proteins in neurons (Grundke-Iqbal, I. et al, "Abnormal Phosphorylation of the Micro tubule-Associated Protein x (tau) in Alzheimer Cytoskeletal Pathology," Proc. Natl. Acad. Sci. U.S.A. 55:4913-4917 (1986)); 2) aberrant expression of genes modulated with neuritic sprouting such as the growth associated protein, GAP-43 (de la Monte, S.M. et al., "Aberrant GAP-43 Gene Expression in Alzheimer's Disease," Am. J. Pathol. 147:934-946 (1995)), constitutive endothelial nitric oxide synthase (de la Monte, S.M. and Bloch, K.D. "Aberrant Expression of the Constitutive Endothelial Nitric Oxide Synthase Gene in <br><br> Alzheimer's Disease," Molecular and Chemical Neuropathy 29: (in press)) transforming growth factor P (Peress, N.S. and Perillo, E., "Differential Expression of TGF-beta 1, 2, and 3 Isotypes in Alzheimer's Disease: a Comparative Immunohistochemical Study With Cerebral Infarction, Aged Human and Mouse Control Brains," J. Neuropathol. Exp. Neurol. 54: 802-811 (1995)), and metallothionine-3 (Aschner, M. "The Functional Significance of Brain Metallothioneins," Faseb. J. 10:1129-1136 (1996)); 3) increased expression of genes associated with glial cell activation, such as glial fibrillary acidic protein (Goodison, K..L. et al., "Neuronal and Glial Gene Expression in Neocortex of Down's Syndrome," J. Neuropathol. Exp. Neurol. 52:192-198 (1993)) and alpha-1 antichymotrypsin (Pasternack, J.M. et al., "Astrocytes in Alzheimer's Disease Gray Matter Express Alpha 1-Antichymotrypsin mRNA," Am. J. Path. 735:827-834(1989); and 4) altered expression of genes that protect neurons from either cytotoxic or programmed cell death, including sulfated glycoprotein-2 (May, P.C. et al., "Dynamics of Gene Expression for aHippocampal Glycoprotein Elevated in Alzheimer's Disease and in Response to Experimental Lesions in Rat," Neuron 5:831-839 (1990), cathepsin D (Cataldo, A.M. et al, "Gene Expression and Cellular Content of Cathepsin D in Alzheimer's Disease Brain: Evidence for Early Up-Regulationofthe Endosomal-Lysosomal System," Neuron 74:671-680 (1995)), superoxide dismutase 1 (Somerville, M.J. et al., "Localization and Quantitation of 68 kDA Neurofilament and Superoxide Dismutase-1 mRNA in Alzheimer Brains, Brain Res. Mol. Brain Res. 9:1-8 (1991), mitochondrial cytochrome oxidase (Chandrasekaran, K. et al., "Impairment in Mitochondrial Cytochrome Oxidase Gene Expression In Alzheimer Disease." Brain Res. Mol. Brain. Res. 24:336-340 (1994)), Clq component of complement (Fischer, B. et al., "Complement Clq and C3 mRNA Expression in the Frontal Cortex of Alzheimer's Patients," J. Mol. Med. 73:465-411 (1995)), Calbindin D28k (Yamagishi, M. et al, "Ontogenetic Expression of Spot 35 Protein (Calbindin-D28k) in Human Olfactory Receptor Neurons and its Decrease in Alzheimer's Disease Patients," Ann. Ontol. Rhinol. Laryngol. 705:132-139 <br><br> (1996), and bcl-2 (O'Barr, S. et al, "Expression of the Protooncogene bcl-2 in Alzheimer's Disease Brain," Neurobiol. Aging 17.131-136 (1996). <br><br> In previous studies, we demonstrated increased immunoreactivity in AD brains using a polyclonal antisera prepared against a pancreatic protein (Ozturk, <br><br> M. et al, "Elevated Levels of an Exocrine Pancreatic Secretory Protein in Alzheimer's Disease Brain," Proc. Natl. Acad. Sci. U.S.A. 86:419-423 (1989); de la Monte, S.M. et al., "Enhanced Expression of an Exocrine Pancreatic Protein in Alzheimer's Disease and the Developing Human Brain," J. Clin. lnvest.86:\0Q4'\b\2 (1990); W090/06993). Using such polyclonal antibodies, <br><br> we isolated the AD7c-NTP cDNA from an AD brain expression library (W094/23756). In W094/23756, this clone is also referred to as AD 10-7, which was deposited in DH1 cells at the ATCC under accession no. 69262. The nucleotide sequence of this cDNA is shown in Fig. 16R of W094/23756. <br><br> However, this sequence comprises numerous errors. See also W096/15272 (Seq. <br><br> ID No. 120, pages 168-170), which also comprises numerous errors. As a result, <br><br> the predicted amino acid sequence (Seq. ID No. 121; W096/15272) is also wrong. <br><br> Summary of the Invention <br><br> The reader's attention is drawn to our related New Zealand Patent No. 337445 (NZ337445). <br><br> NZ337445 is related to transgenic animals and cell lines which over express the AD7c-NTP and use thereof to screen candidate drugs for use in the treatment or prevention of Alzheimer's disease, neuroectodermal tumors, <br><br> malignant astrocytomas and glioblastomas. <br><br> In particular, NZ337445 relates to a DNA construct, wherein said DNA construct comprises a DNA molecule having Seq. ID No. 1 or a DNA sequence at least 40% homolous thereto, or a fragment thereof, wherein the DNA molecule is under control of a heterlogous, neuro-specific promoter. <br><br> NZ337445 also relates to cell lines containing the said DNA construct. <br><br> " intellectual property office of n.z <br><br> 2 6 AUG 20M <br><br> Dcpcu'cri <br><br> NZ337445 relates to transgenic non-human animal, all of whose germ and somatic cells comprises molecule of Seq. ED No. 1 or a DNA molecule which is at least 40% homologous thereto. Preferably, the transgenic animals over-express AD7c-NTP. <br><br> NZ337445 also relates to an in vitro method for screening candidate drugs that are potentially useful for the treatment or prevention of Alzheimer's disease, neuroectodermal tumors, malignant astrocytomas, and glioblastomas, <br><br> which comprises <br><br> (a) contacting a candidate drug with a host transfected with a DNA construct, wherein the DNA construct comprises a DNA molecule of Seq. ID No. <br><br> 1 or a DNA molecule at least 40% homologous thereto, or a fragment thereof, and wherein said host over expresses the protein coded for by said DNA molecule, and <br><br> (b) detecting at least one of the following: <br><br> (i) the suppression or prevention of expression of the protein; <br><br> (ii) the increased degradation of the protein; or <br><br> (iii) the reduction of frequency of at least one of neuritic sprouting, nerve cell death, <br><br> degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons in the host; <br><br> due to the drug candidate compared to a control host that has not received the candidate drug. <br><br> In a preferred embodiment of NZ337445, the host is a transgenic animal. In another preferred embodiment, the host is a cell in vitro. <br><br> Accordingly, in one aspect NZ337445 provides an in vivo method for screening a candidate drug that is potentially ... <br><br> useful for the treatment or prevention of Alzheimer's disease, neuroectodermal tumors, malignant astrocytomas, and glioblastomas, which comprises <br><br> (a) administering a candidate drug to the transgenic animal of the invention, <br><br> and <br><br> (b) detecting at least one of the following: <br><br> (i) the suppression or prevention of expression of the protein coded for by the DNA construct contained by said animal; <br><br> INTELLECTUAL PROPERTY OFFICE OF N.Z <br><br> 2 6 AUG 2004 <br><br> RECEIVED <br><br> -6a- <br><br> (TO the Increased degradation ofthe protein coded for by the <br><br> DNA construct contained by said animal; or (iii) the reduction of frequency of at least one of neuritic sprouting, nerve cell death, <br><br> degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons in the host; <br><br> due to the drug candidate compared to a control animal which has not received the candidate drug. <br><br> NZ337445 is also directed to antisense oligonucleotides which are complementary to an NTP nucleic acid sequence and which is nonhomologous to PTP nucleic acid sequences and that correspond to regions that were incorrectly sequenced in the past, as well as pharmaceutical compositions comprising such oligonucleotides and a pharmaceutically acceptable carrier. <br><br> NZ337445 is also directed to ribozymes comprising a target sequence which is complementary to an NTP sequence and nonhomologous to PTP nucleic acid sequences and that correspond to regions that were incorrectly sequenced in intellectual property office of n.z <br><br> 2 6 AUG 2004 <br><br> Bcrcu^cn <br><br> the past, as well as pharmaceutical compositions comprising such ribozymes and a pharmaceutical^ acceptable carrier. <br><br> NZ337445 is also directed to oligodeoxynucleotides that form triple stranded regions with the AD7c-NTP gene, which are nonhomologous to PTP nucleic acid sequences, and that correspond to regions that were incorrectly sequenced in the past, as well as pharmaceutical compositions comprising such oligodeoxynucleotides and a pharmaceutically acceptable carrier. ■' - <br><br> Described but not claimed is a method of achieving pharmaceutical delivery of the antisense oligonucleotides, ribozymes and triple helix oligonucleotides to the brain through acceptable carriers or expression vectors. <br><br> The present invention is directed to the therapeutic use of the antisense oligonucleotides, ribozymes and triple helix oligonucleotides to modify or improve dementias of the Alzheimer's type of neuronal degeneration; as well as to treatof prevent neuroectodermal tumors, malignant astrocytomas, and glioblastomas. <br><br> More specifically, in one aspect the present invention provides a method to treat or prevent dementias of the Alzheimer's type of neuronal degeneration; or to treat or prevent neuroectodermal tumors, malignant astrocytomas, or glioblastomas, comprising administering to a non-human animal in need thereof an antisense oligonucleotide, a ribozyme or a triple helix-forming oligonucleotide selected from <br><br> (a) an antisense oligonucleotide which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l; <br><br> (b) a ribozyme comprising a target sequence which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l; <br><br> (c) an oligodeoxynucleotide that forms triple stranded regions with the a region of AD7c-NTP coding nucleic acid and having the sequence 3'X5'-L-5'X3', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq. ID No.l, and wherein L represents an oligonucleotide linker or a bond; and <br><br> "intellectual property office of n.z <br><br> 2 6 AUG 200'! <br><br> RECEIVED <br><br> 00 01 <br><br> -7A- <br><br> intellectual property office of n.z. <br><br> 2 1 FEB 2002 received <br><br> (d) an oligodeoxynucleotide that forms triple stranded regions with a region of AD7c-NTP coding nucleic acid and having the sequence 5'X3'-L-3'X5', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq ID No.l, and wherein L represents an oligonucleotide linker or a bond. <br><br> In a further aspect, the present invention provides a use, in the preparation of a medicament for treating or preventing a condition selected from the group comprising dementias of the Alzheimer's type of neuronal degeneration, neuroectodermal tumors, malignant astrocytomas, and glioblastomas in a patient in need thereof, of an antisense oligonucleotide, a ribozyme or a triple helix-forming oligonucleotide selected from <br><br> (a) an antisense oligonucleotide which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l; <br><br> (b) a ribozyme comprising a target sequence which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l; <br><br> (c) an oligodeoxynucleotide that forms triple stranded regions with the a region of AD7c-NTP coding nucleic acid and having the sequence 3'X5'-L-5'X3', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq. ID No.l, and wherein L represents an oligonucleotide linker or a bond; and <br><br> (d) an oligodeoxynucleotide that forms triple stranded regions with a region of AD7c-NTP coding nucleic acid and having the sequence 5'X3'-L-3'X5', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq ID No.l, and wherein L represents an oligonucleotide linker or a bond. <br><br> -7B- <br><br> 2 1 FEB 2002 received <br><br> Brief Description of the Figures v <br><br> Fig. 1 depicts the nucleotide and translated amino acid sequence (Seq ID Nos. 1 and 2) of the AD7c-NTP cDNA. The shaded region corresponds to the nucleic acid sequences detected in 6 AD brains by RT-PCR analysis of mRNA. The cDNA exhibits significant homology with Alu gene, and to an unknown gene in the Huntington region, Chromosome 4ql6.3 (underlined). The open reading frame begins with the first methionine codon. The translated amino acid sequence encodes a 41.3 kD protein with a hydrophobic leader sequence (italics) followed by a myristoylation motif (bold, italics) and potential Al cleavage site. That same region (italics, underlined) exhibits significant homology with the insulin/IGF-1 chimeric receptor. There are 17 potential glycogen synthase kinase-3, protein kinase C, or cAMP or Ca-dependent kinase II phosphorylation motifs and one transforming growth factor (tgf) motif (double underlined). The embolded amino acid sequences exhibit significant homology with the A4 alternatively spliced <br><br> mutant form ofNF2, P subunit of integrin, and human decay accelerating factor 2 precursor. The boxed amino acid sequences exhibit significant homology with human integral membrane protein and myelin oligoglycoprotein-16. <br><br> Figs. 2A-2D depict AD7c-NTP expression in vitro and in vivo. (2A): Recombinant protein detected by in vitro translation using sense strand cRNA transcripts. (2B): Western blot analysis of purified recombinant protein demonstrating specific immunoreactivity with the Tag and N3I4 AD7c-NTP monoclonal antibodies, but not with non-relevant FB50 monoclonal antibody. (2C): Western blot analysis of BOSC cells stably transfected with pcDNA3-AD7c-NTP or pcDNA3 (empty vector). The blots were probed with the N3I4 AD7c-NTP antibody. (2D): Significantly increased levels of the 41-45 kD AD7c-NTP protein in AD frontal lobe relative to age-matched control frontal lobe tissue. Similar results were obtained for temporal lobe tissue. (2E): Higher levels of the 41-45 kD and 19-21 kD AD7c-NTP proteins in late, end-stage (L) AD compared with early, less symptomatic (E) AD. All tissue samples were taken from the frontal lobe. Note the clusters of 3 or 4 bands between ~41 and -45 kD, probably corresponding to different degrees of phosphorylation. (2F): Western blot analysis of postmortem ventricular fluid demonstrating higher levels of the -41 kD AD7c-NTP molecules in AD compared with aged control samples using the N3I4 antibody. The ~28-30 kD band may represent a degradation product. Also note detection of the -19-21 kD N3I4-immunoreactive molecules in AD. <br><br> Figs. 3A-3F depict AD7c-NTP mRNA expression in AD and aged control brains. Northern blot analysis of AD and aged control frontal lobe RNA detected -1.4 kB transcripts corresponding to the size of the AD&amp;c-NTP cDNA. In addition, -0.9 kB transcripts corresponding to a different cDNA were detected in all brains, but not in other tissues. Densitometric analysis of the autoradiograms revealed variable levels of AD7c-NTP mRNA expression in the AD group (3 A), but significantly higher mean levels of the 1.4 kB AD7c-NTP transcript in the AD (N=17) relative to the aged control (N=l 1) group (P&lt;0.01). (Figs. 3C and 3D): Brightfield photomicrographs of in situ hybridization results using antisense (3C) <br><br> or sense (3D; negative control) digoxigenin-labeled cRNA probes. Arrows indicate examples of neurons and dark grains represent positive hybridization signals. (Figs. 3E and 3F): Darkfield photomicrographs of in situ hybridization results demonstrating more intense labeling (white grains) in AD (3E) relative to aged control (3F) cortical neurons (arrows) in the frontal lobe. Probe labeling was detected with antidigoxigenin and alkaline phosphatase substrates (see below). The white signals aggregated over neurons (pyramidal shaped) represent positive results, and black areas indicate absent probe binding. <br><br> Figs. 4A-4H depict increased AD7c-NTP immunoreactivity in AD (4A) relative to aged control (4B) conical neurons by immunohistochemical staining with the N2T8 (4A and 4B). N2J1 immunoreactivity in AD brains (Figs. 4C, 4E-4H) demonstrating high-level AD7c-NTP expression or accumulation in the perikarya of cytologically intact (4C) as well as degenerating (4E) neurons. In addition, the N2J1 antibody was immunoreactive with abnormal dystrophic cell processes occurring in aggregates (sprouts) (4F), dispersed in the white matter (4G), and corresponding to irregular beaded axons (4H). Fig. 4D depicts AD cerebral cortex immunostained with non-relevant antibody. The sections in Figs. 4A and 4B were counter stained with hematoxylin to provide a contrasting background. <br><br> Figs. 5A and 5B depict graphs showing increased cell death in pcDNA3-AD7c-NTP transfected SH-Sy5y cells. Synchronized cells were fed with medium containing 10% fetal calf serum, and DNA synthesis was assessed by 3H-thymidine incorporation into DNA (5A). The density of viable cells was determined at each time point (5 A). Despite higher levels of DNA synthesis (5B). cell density was significantly reduced in 4 replicate AD7c-NTP-transfected cultures compared with control (pcDNA3-transfected) cells. AD7c-NTP-transfected cells also exhibited increased nuclear p53 immunoreactivity and increased nuclear DNA fragmentation by the in situ assay for nicked DNA (TUNEL), suggesting that over-expression of AD7c-NTP in neuronal cells causes apoptosis. <br><br> -10- <br><br> Figs. 6A-6G show that AD7c-NTP over-expression in transfected neuronal cells results in increased neuritic sprouting. (6A): SH-Sy5y cells stably transfected with pcDNA3 (empty vector). (6B-6D): SH-Sy5y cells stably transfected with pcDNA3-AD7c-NTP. Note fine neuritic processes (arrows) on most cells in Figs. 6B-6D. Also note lower cell density and numerous round refractile dead cells (arrowheads) in Fig. 6D compared with Fig. 6A. (Fig. 6E-6G): Imrnunocytochemical staining of SH-Sy5y cells stably transfected with pcDNA3 (6E) or pcDNA3-AD7c-NTP (6F, 6G) using N3I4 monoclonal antibody. Note intense labeling of perikarya and cell processes (arrows) in 6F and 6G and absent labeling in 6E. <br><br> Figs. 7A-7C depict modulation of gene expression following IPTG induction of AD7c-NTP expression. LacA-control cells (Fig. 7A) lack AD7c-NTP; LacB-B6 cells (Fig. 7B) and LacF-B6 cells (Fig. 7C) are two different clones with different levels of AD7c-NTP induction. Changes in the level of expression 24 hours after induction are indicated for genes involved in AD, neural sprouting, and apoptosis. <br><br> Figs. 8 A-8D depict IPT G dose-dependent increases in the level of the NTP (Fig. A), Tau (Fig. B), Synaptophysin (Fig. C) and p53 (Fig. D) proteins. The percent change of the amount of each protein is presented as a function of IPTG concentration (mM). <br><br> Figures 9A and 9B depict the effects of AD7c-NTP expression in CYZ neuronal cells on metabolic (MTT) activity and cell viability. Lac A-Lac F represent six different clones, and B6 indicates AD7c-NTP expression. The percent change for MTT activity (Fig. 9A) and cell viability (Fig. 9B) are indicated for control (Lac A-Control) and AD7c-NTP expressing cell lines. <br><br> Figures 10A and 10B depict the effect of AD7c-NTP expression on cell viability. Nonexpressing Lac A Control and cell lines expressing AD7c-NTP at various levels. Lac B-B6 and Lac F-B6, were assayed for viability after varying exposure time to the protein expression inducing agent (IPTG) and various oxidative stress toxins. <br><br> Figure 11 depictsDNA fragmentation after IPTG induction of AD7c-NTP expression, thereby providing a quantitative assessment of apoptosis. Stably transfected CYZ neuronal cell lines, Lac A, Lac B and Lac F, which express various levels of AD7c-NTP after induction, were incubated in the presence of 32dCTP label. The amount of radioactive isotope incorporated, under control (uninduced) and IPTG induction conditions, into the respective cell line DNAs is presented. <br><br> Figure 12 depicts the percent change in viability for cells stably transfected with and expressing AD7c-NTP under conditions that promote and reduce or block oxidative stress. Agents promoting oxidative stress are the following: hydrogen peroxide (H:0:) diethyldithiocarbamic acid (DDC), S-nitro-N-acetyl-penicillamine (SNAP) and N-acetyl cysteine; and the agent utilized to block or reduce oxidative stress is pygroglutamate (PG). <br><br> Detailed Description of the Preferred Embodiments Definitions <br><br> In the description that follows, a number of terms used in recombinant DNA technology are utilized extensively. In order to provide a clear and consistent understanding of the specification and claims, including the scope to be given such terms, the following definitions are provided. <br><br> Cloning vector. A plasmid or phage DNA or other DNA sequence which is able to replicate autonomously in a host cell, and which is characterized by one or a small number of restriction endonuclease recognition sites at which such DNA sequences may be cut in a determinable fashion without loss of an essential biological function of the vector, and into which a DNA fragment may be spliced in order to bring about its replication and cloning. The cloning vector may further contain a marker suitable for use in the identification of cells transformed with the <br><br> cloning vector. Markers, for example, provide tetracycline resistance or ampicillin resistance. <br><br> Expression vector. A vector similar to a cloning vector but which is capable of enhancing the expression of a gene which has been cloned into it, after transformation into a host. The cloned gene is usually placed under the control of (i.e., operably linked to) certain control sequences such as promoter sequences. Promoter sequences may be either constitutive or inducible. <br><br> Substantially pure. As used herein means that the desired purified protein is essentially free from contaminating cellular components, said components being associated with the desired protein in nature, as evidenced by a single band following polyacrylamide-sodium dodecyl sulfate gel electrophoresis. Contaminating cellular components may include, but are not limited to, proteinaceous, carbohydrate, or lipid impurities. <br><br> The term "substantially pure" is further meant to describe a molecule which is homogeneous by one or more purity or homogeneity characteristics used by those of skill in the art. For example, a substantially pure NTP will show constant and reproducible characteristics within standard experimental deviations for parameters such as the following: molecular weight, chromatographic migration, amino acid composition, amino acid sequence, blocked or unblocked N-terminus, HPLC elution profile, biological activity, and other such parameters. The term, however, is not meant to exclude artificial or synthetic mixtures of the factor with other compounds. In addition, the term is not meant to exclude NTP fusion proteins isolated from a recombinant host. <br><br> Recombinant Host. According to the invention, a recombinant host may be any prokaryotic or eukaryotic host cell which contains the desired cloned genes on an expression vector or cloning vector. This term is also meant to include those prokaryotic or eukaryotic cells that have been genetically engineered to contain the desired gene(s) in the chromosome or genome of that organism. For examples of such hosts, see Sambrook et al.. Molecular Cloning: A Laboratory Manual. Second Edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, <br><br> -13- <br><br> New York (1989). Preferred recombinant hosts are neuronal cells transformed with the DNA construct of the invention. Such neuronal cells include brain cells that have been isolated after mechanical disassociation of an animal brain or other available neuronal cell lines. <br><br> Recombinant vector. Any cloning vector or expression vector which contains the desired cloned gene(s). <br><br> Host Animal. Transgenic animals, all of whose germ and somatic cells contain the DNA construct of the invention. Such transgenic animals are in general vertebrates. Preferred Host Animals are mammals such as non-human primates, mice, sheep, pigs, cattle, goats, guinea pigs, rodents, e.g. rats, and the like. The term Host Animal also includes animals in all stages of development, including embryonic and fetal stages. <br><br> Promoter. A DNA sequence generally described as the 5' region of a gene, located proximal to the start codon. The transcription of an adjacent gene(s) is initiated at the promoter region. If a promoter is an inducible promoter, then the rate of transcription increases in response to an inducing agent. In contrast, the rate of transcription is not regulated by an inducing agent if the promoter is a constitutive promoter. According to the invention, preferred promoters are heterologous to the AD7c-NTP gene, that is. the promoters do not drive expression of the gene in a human. Such promoters include the CMV promoter (InVitrogen, San Diego, CA), the SV40, MMTV, and hMTIIa promoters (U.S. 5,457,034), the HSV-1 4/5 promoter (U.S. 5,501,979), and the early intermediate HCMV promoter (W092/17581). Also, it is preferred that the promoter is neuro-specific, that is, it is induced selectively in neuronal tissue. Also, neuro-specific enhancer elements may be employed. Examples of neuro-specific promoters include but are not limited to the promoter which controls the neurofilament gene (W091/02788; Byme and Ruddle, Proc. Natl. Acad. Sci. USA 86:5473-5477 (1989)), the neuron specific promoter of the human neurofilament light gene (NFL) (U.S. 5,569.827); the promoter of the P2-subunit of the neuronal nicotinic acetylcholine receptor (EP 0 171 105; U.S. appl. no. <br><br> -14- <br><br> 08/358,627), the hThy-1 promoter (W095/03397; U.S. appl. no. 08/096,944; Gordon, J. et al., Cell 50:445-452 (1987)); the Tal a-tubulin promoter (W095/25795; U.S. appl. no. 08/215,083; Gloster et al, J. Neurosci. 74:7319-7330 (1994)), the APP promoter, the rat neuron specific promoter, the human p actin gene promoter, the human platelet derived growth factor B (PDGF-B) chain gene promoter, the rat sodium channel gene promoter, the mouse myelin basic protein gene promoter, the human copper-zinc superoxide dismutase gene promoter, mammalian POU-domain regulatory gene promoter (W093/14200: U.S. appl. nos. 07/817,584 and 07/915,469); human platelet derived growth factor B (PDGF-B) chain gene promoter (W096/40895: U.S. appl. nos. 08/486,018 and 08/486,538; Sasahara et al, Cell 64:2X1-22.1 (1991)); and the neuron-specific enolase promoter (McConiogue et al. Aging 15:S12 (1994); Higgins et al, Ann Neurol. 55:598-607 (1995); Mucke et al, Brain Res. 666:151-161 (1994); Higgins et al, Proc. Natl. Acad. Sci USA 92:4402-4406 (1995); W096/40896; U.S. appl. no. 08/480,653; and U.S. 5,387,742); and sequences that regulate the oligodendroglial-specific expression of JC virus, glial-specific expression of the proteolipid protein, and the glial fibrillary acidic protein genes (U.S. Patent No. 5,082,670). Other neuro-specific promoters will be readily apparent to those of skill in the art. Since protein phosphorylation is critical for neuronal regulation (Kennedy, "Second Messengers and Neuronal Function," in An Introduction to Molecular Neurobiology, Hall, Ed., Sinauer Associates, Inc. (1992)), protein kinase promoter sequences can be used to achieve sufficient levels of NTP gene expression. <br><br> Gene. A DNA sequence that contains information needed for expressing a polypeptide or protein. <br><br> Structural gene. A DNA sequence that is transcribed into messenger RN A (mRNA) that is then translated into a sequence of amino acids characteristic of a specific polypeptide. <br><br> Antisense RNA gene/Antisense RNA. In eukaryotes, mRNA is transcribed by RNA polymerase II. However, it is also known that one may <br><br> construct a gene containing a RNA polymerase II template wherein a RNA sequence is transcribed which has a sequence complementary to that of a specific mRNA but is not normally translated. Such a gene construct is herein termed an "antisense RNA gene" and such a RNA transcript is termed an "antisense RNA." Antisense RNAs are not normally translatable due to the presence of translation stop codons in the antisense RNA sequence. <br><br> Antisense oligonucleotide. A DNA or RNA molecule or a derivative of a DNA or RNA molecule containing a nucleotide sequence which is complementary to that of a specific mRNA. An antisense oligonucleotide binds to the complementary sequence in a specific mRNA and inhibits translation of the mRNA. There are many known derivatives of such DNA and RNA molecules. See, for example, U.S. Patent Nos. 5,602,240, 5,596,091, 5,506,212, 5,521,302. 5,541,307, 5,510,476, 5,514,787, 5,543,507, 5,512,438, 5,510,239, 5,514,577. 5,519,134, 5,554,746,5,276,019,5,286,717, 5,264,423, as well as W096/35706, W096/32474, W096/29337 (thiono triester modified antisense oligodeoxynucleotide phosphorothioates), W094/17093 (oligonucleotide alkylphosphonates and alkylphosphothioates), W094/08004 (oligonucleotide phosphothioates, methyl phosphates, phosphoramidates, dithioates. bridged phosphorothioates. bridge phosphoramidates. sulfones. sulfates, ketos, phosphate esters and phosphorobutylamines (van der Krol et al.. Biotech. (5:958-976 (1988); Uhlmann et al, Chem. Rev. 90:542-585 (1990)), W094/02499 (oligonucleotide alkylphosphonothioates and arylphosphonothioates). and W092/20697 (3'-end capped oligonucleotides). Particular NTP antisense oligonucleotides of the present invention include derivatives such as S-oIigonucleotides (phosphorothioate derivatives or S-oligos, see, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press (1989)). S-oligos (nucleoside phosphorothioates) are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom. The S-oligos of the present invention may be prepared by treatment of the corresponding O-oligos with 3H-l,2-benzodithiol-3-one-1,1 -dioxide which is a <br><br> -16- <br><br> sulfur transfer reagent. See Iyer et al., J. Org. Chem. 55:4693-4698 (1990); and Iyer et al., J. Am. Chem. Soc. 7/2:1253-1254 (1990). <br><br> Antisense Therapy. A method of treatment wherein antisense oligonucleotides are administered to a patient in order to inhibit the expression of the corresponding protein. <br><br> Complementary DNA (cDNA). A "complementary DNA," or "cDNA" gene includes recombinant genes synthesized by reverse transcription of mRNA and from which intervening sequences (introns) have been removed. <br><br> Expression. Expression is the process by which a polypeptide is produced from a structural gene. The process involves transcription of the gene into mRNA and the translation of such mRNA into polypeptide(s). <br><br> Homologous/Nonhomologous Two nucleic acid molecules are considered to be "homologous" if their nucleotide sequences share a similarity of greater than 40%, as determined by HASH-coding algorithms (Wilber, W.J. and Lipman. D.J., Proc. Natl. Acad. Sci. 80:126-120 (1983)). Two nucleic acid molecules are considered to be "nonhomologous" if their nucleotide sequences share a similarity of less than 40%. <br><br> Ribozyme. A ribozyme is an RNA molecule that contains a catalytic center. The term includes RNA enzymes, self-splicing RNAs, and self-cleaving RNAs. <br><br> Ribozyme Therapy. A method of treatment wherein ribozyme is administered to a patient in order to inhibit the translation of the target mRNA. <br><br> Fragment. A "fragment" of a molecule such as NTP is meant to refer to any polypeptide subset of that molecule. <br><br> Functional Derivative. The term "functional derivatives" is intended to include the "variants," "analogues," or "chemical derivatives" of the molecule. A "variant" of a molecule such as NTP is meant to refer to a naturally occurring molecule substantially similar to either the entire molecule, or a fragment thereof. An "analogue" of a molecule such as NTP is meant to refer to a non-natural molecule substantially similar to either the entire molecule or a fragment thereof. <br><br> -17- <br><br> A molecule is said to be "substantially similar" to another molecule if the sequence of amino acids in both molecules is substantially the same, and if both molecules possess a similar biological activity. Thus, provided that two molecules possess a similar activity, they are considered variants as that term is used herein even if one of the molecules contains additional amino acid residues not found in the other, or if the sequence of amino acid residues is not identical. <br><br> As used herein, a molecule is said to be a "chemical derivative" of another molecule when it contains additional chemical moieties not normally a part of the molecule. Such moieties may improve the molecule's solubility, absorption, biological half-life. etc. The moieties may alternatively decrease the toxicity of the molecule, eliminate or attenuate any undesirable side effect of the molecule, etc. Examples of moieties capable of mediating such effects are disclosed in Remington's Pharmaceutical Sciences (1980) and will be apparent to those of ordinary skill in the art. <br><br> AD7c-NTP. The term " AD7c-NTP" refers to the protein having sequence ID No. 2 as well as allelic variants thereof. <br><br> We have isolated a cDNA designated AD7c-NTP, that is expressed in neurons, and over-expressed in brains with AD. The 1442-nucleotide AD7c-NTP cDNA encodes a ~41 kD membrane spanning protein that has a hydrophobic leader sequence and myristy lation motif near the amino terminus. The AD7c-NTP cDNA is an Alu sequence-containing gene with three regions of significant homology to the alternatively spliced A4 form ofNF2, the P1 subunit of integrin, human integral membrane protein, myelin oligoglycoprotein-16 precursor, and human decay accelerating factor 2 precursor, and two regions with significant homology with sequences in the Huntington's disease region on Chromosome 4pl6.3. Expression of AD7c-NTP was confirmed by nucleic acid sequencing of RT-PCR products isolated from brain. AD7c-NTP cRNA probes hybridized with 1.4 kB and 0.9 kB mRNA transcripts by Northern blot analysis, and monoclonal antibodies generated with the recombinant protein were immunoreactive with -39-45 kD and -19-21 kD molecules by Western blot analysis of human brain. <br><br> -18- <br><br> Quantitation of data obtained from 17 AD and 11 age-matched control brains demonstrated significantly higher levels of AD7c-NTP expression in AD. In situ hybridization and immunostaining studies localized AD7c-NTP gene expression in neurons, and confirmed the over-expression associated with AD neurodegeneration. Increased AD7c-NTP protein levels were also detectable in cerebrospinal fluid by Western blot analysis. The results suggest that abnormal AD7c-NTP gene expression is associated with AD neurodegeneration. Thus, <br><br> abnormal expression of AD7c-NTP is a phenotype associated with Alzheimer's disease. <br><br> The confirmation that AD7c-NTP expression leads to Alzheimer's disease led to the expectation that transgenic animals and cell lines which over express the AD7c-NTP can be used to screen drugs for use in the treatment or prevention of Alzheimer's disease, neuroectodermal tumors, malignant astrocytomas and glioblastomas. <br><br> NZ337445 relates to a DNA construct, wherein said DNA construct comprises a DNA molecule of Seq. ID No. 1, or a fragment thereof, or a DNA molecule which is at least 40% homologous thereto, more preferably, at least 85% <br><br> homologous thereto, most preferably, at least 90% homologous thereto. <br><br> Preferably, the DNA construct encodes AD7c-NTP having Seq. ID No. 2. Also preferably, the DNA sequence is under control of a heterologous neuro-specific promoter. Examples of promoters that can be used to drive expression of AD7c-NTP in a host cell are described above. Having the promoter in hand, one may simply ligate the promoter to the DNA molecule of Seq. ID No. 1. Methods for ligating DNA fragments are well known to those of ordinary skill in the art. <br><br> Preferably, the DNA molecule having Seq. ID No. 1 is ligated to a plasmid which contains the promoter and which results in the promoter being in operable linkage to the AD7c-NTP DNA sequence. <br><br> Fragments of the DNA molecule of NZ337445 code for proteins having the activity of AD7c-NTP, that is, the DNA fragments induce neutitic sprouting, <br><br> nerve cell death, nerve cell degeneration, neurofibrillary tangles, and/or irregular ntellectual property office of n.z <br><br> 2 6 AUG 2004 <br><br> RECEIVED <br><br> /I <br><br> -19- <br><br> intellectual property office of n.z, <br><br> 2 1 FEB 2002 received swollen neurites in a host which expresses the fragment. Such hosts include cellular hosts and transgenic animals. <br><br> DNA molecules which are at least 40%, 85% or 90% homologous to Seq. ID No. 1 may be isolated from cDNA libraries of humans and animals by hybridization under stringent conditions to the DNA molecule of Seq. ID No. 1 according to methods known to those of skill in the art. Stringent hybridization conditions are employed which select for DNA molecules having at least 40%, 85% and 90% homology to Seq. ID No. I are described in Sambrook et al., In: Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989); and Maniatis et al., Molecular Cloning - A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1985. The hybridizations may be carried out in 6 x SSC/5 x Denhardt's solution/0.1% SDS at 65°C. The degree of stringency is determined in the washing step. Thus, suitable conditions include 0.2 x SSC/0.01 % SDS/65 °C and 0.1 x SSC/0.01% SDS/65 °C. <br><br> NZ337445 also relates to cells containing the said DNA construct. <br><br> Examples of suitable cells that may contain the DNA construct include eukaryotic and prokaryotic cells. Preferred are eukaryotic cells such as those derived from a vertebrate animal including human cells, non-human primate cells, porcine cells, ovine cells and the like. Further, it is contemplated that the cell line may be a neuronal cell line from one of these vertebrate animals. Examples of such cell lines include SH-Sy5y, pNET-1, pNET-2, hNTs (Stratagene, Inc.), and A172 (ATCC) neuronal cells. See O'Barr, S. et al., Neurobiol. Aging 77:131-136 (1996); Ozturk, M. et al, Proc. Natl. Acad. Sci. USA 86:419-423 (1989); Bieldler, et al, Cancer Res. 33:2643-2652 (1973); and The et al. Nature Genet. 5:2643-2652 (1993). <br><br> Methods for introducing DNA constructs into cells in vitro, in vivo and ex vivo are well known to those of ordinary skill in the art. See, for example, U.S. patent nos. 5,595,899, 5,521,291, 5,166,320, 5,547,932, 5,354,844, 5,399,346, W094/10569 and Citron et al. Nature 360:622-674 (1995). <br><br> K 0 Q H 1" Pi <br><br> 3^0 <br><br> V <br><br> -20- <br><br> intellectual property office of n.z. <br><br> 2 1 FEB 2002 received <br><br> NZ337445 also relates to transgenic non-human animals which comprise the DNA construct of the invention in each of its germ and somatic cells and which over express AD7c-NTP. Such transgenic animals may be obtained, for example, by injecting the DNA construct of the invention into a fertilized egg which is allowed to develop into an adult animal. To prepare a transgenic animal, a few hundred DNA molecules are injected into the pro-nucleus of a fertilized one cell egg. The micro injected eggs are then transferred into the oviducts of pseudopregnant foster mothers and allowed to develop. It has been reported by Brinster et al., Proc. Natl. Acad. Sci. USA 82:4438-4442 (1985), that about 25% of mice which develop will inherit one or more copies of the micro injected DNA. Alternatively, the transgenic animals may be obtained by utilizing recombinant ES cells for the generation of the transgenes, as described by Gossler et al., Proc. Natl. Acad. Sci. USA 83:9065-9069 (1986). The offspring may be analyzed for the integration of the transgene by isolating genomic DNA from tail tissue and the fragment coding for AD7c-NTP identified by conventional DNA-hybridization techniques (Southern, J. Mol. Biol. 9£:503-517 (1975)). Animals positive for the AD7c-NTP gene are further bred to expand the colonies of AD7c-NTP mice. General and specific examples of methods of preparing transgenic animals are disclosed in U.S. 5,602,299, 5,366,894. 5,464,758, 5,569,827, W096/40896 (U.S. appl. no. 08/480,653); W096/40895 (U.S. appl. nos. 08/486,018 and 08/486,536); W093/14200 (U.S. appl. nos. 07/817,584 and 07/915,469); W095/03397 (U.S. appl. no. 08/096,944); W095/25792 (U.S. appl. no. 08/215,083); EP 0 717 105 (U.S. appl. no. 08/358,627); and Hogan et al.. Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1986); Hammer et al., Cell 63:1099-1112 (1990). <br><br> Once obtained, the transgenic animals which contain the AD7c-NTP may be analyzed by immunohistology for evidence of AD7c-NTP expression as well as for evidence of neuronal or neuritic abnormalities associated with Alzheimer's disease, neuroectodermal tumors, malignant astrocytomas and glioblastomas. <br><br> y y <br><br> -21- <br><br> intellectual property office of n.z. <br><br> 2 1 FEB 2002 received <br><br> Sections of the brains may be stained with antibodies specific for AD7c-NTP, <br><br> either monoclonal or polyclonal. <br><br> NZ337445 also relates to an in vitro method for screening candidate drugs that are potentially useful for the treatment or prevention of Alzheimer's 5 disease, neuroectodermal tumors, malignant astrocytomas, and glioblastomas, <br><br> which comprises <br><br> (a) contacting a candidate drug with a host transfected with a DNA construct, wherein the DNA construct comprises a DNA molecule of Seq. ID No. 1 or a DNA molecule that is at least 90% homologous thereto, and wherein said <br><br> 10 host over expresses the protein coded for by said DNA molecule, and <br><br> (b) detecting at least one of the following: <br><br> (i) the suppression or prevention of expression of the protein; <br><br> (ii) the increased degradation of the protein; or <br><br> (iii) the reduction of frequency of at least one 15 of neuritic sprouting, nerve cell death, <br><br> degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons in the host; <br><br> due to the drug candidate. <br><br> In a preferred embodiment of NZ337445, the host is a transgenic animal. In another preferred embodiment, the host is a cell in vitro. The suppression or prevention of expression, and the increased degradation of the protein such as AD7c-NTP may be detected with antibodies specific for AD7c-NTP. Monoclonal and polyclonal antibodies which are specific for AD7c-NTP as well as methods for the 25 qualitative and quantitative detection of AD7c-NTP are described herein as well as in W094/23756 and U.S. appl. no. 08/340,426. Such testing may be carried out on CSF of the transgenic animal or by immunohistochemical staining of a tissue section from the brain of the animal. In addition, such testing may be carried out by Western blot analysis, ELISA or RIA. <br><br> intellectual property office of n.z. <br><br> 2 1 FEB 2002 <br><br> received <br><br> -22- <br><br> Immunohistochemical staining may also be carried out to determine the frequency of at least one of neuritic sprouting, nerve cell death, degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons in the animal. Since in general the animal will have to be sacrificed, a pool of test and control transgenic animals should be tested. After sacrifice, the relative frequency of neuritic sprouting, nerve cell death, degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons is determined for both groups. If the test group exhibits a reduced frequency of neuritic sprouting, nerve cell death, degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons, the drug may be considered promising for the treatment or prevention of Alzheimer's disease, neuroectodermal tumors, malignant astrocytomas, or glioblastomas. <br><br> When the host is a transgenic animal, the effect of a drug candidate may also be tested by behavioral tests which are designed to assess learning and memory deficits. An example of such a test is the Morris water maze disclosed by Morris, Learn Motivat. 12:239-260 (1981) and W096/40895. <br><br> In the practice of the method of NZ337445, the candidate drug is administered to the transgenic animals or introduced into the culture media of cells derived from the animals or cells transfected with the DNA construct of the invention. The candidate drug may be administered over a period of time and in various dosages, and the animals or animal cells tested for alterations in AD7-c NTP expression, nerve cell degradation or histopathology. In case of transgenic animals, they may also be tested for improvement in behavior tests. <br><br> When cells are to be tested in vitro for the effect of the candidate drug, they are grown in a growth conducive medium and the medium replaced with a media containing the candidate drug. Wide varieties of medias which promote growth of practically any cell type are commercially available, for example, from Life Technologies, Inc. (Gaithersburg, MD). If the candidate drug is only sparingly soluble in the media, a stock solution may be prepared in dimethyl sulfoxide (DMSO). The DMSO solution is then admixed with the media. <br><br> Preferably, the DMSO concentration in the media does not exceed 0.5%, preferably, 0.1%. The cells are then incubated in the presence of the drug-containing media for a preselected time period (e.g. 2-10 hours) at a preselected temperature, for example, about 37°C. At the end of this time period, the media may again be removed and fresh media containing the candidate drug is added. The cells are then incubated for a second preselected time period (e.g. 2-16 hours). This procedure can be repeated as necessary to achieve a significant result. <br><br> After the treatment period, the cells are tested either for the level of NTP expression and/or, if the cells are neuronal cells, examined for the presence and/or frequency of neuritic sprouting, nerve cell death, degenerating neurons, neurofibrillary tangles, or irregular swollen neurites and axons. In order to test for the level of NTP expression, immunohistochemical staining may be carried out as described in the Examples. Alternatively, the plates containing the cells may be centrifuged to pellet cellular debris from the medium, and a sample of the media tested for the NTP concentration. The concentration of NTP may be determined by ELISA with an antibody which is specific for NTP. Methods for carrying out such assays are disclosed in W094/10569 and are well known to those of ordinary skill in the art. The concentration of NTP in the test cells/media is then compared to the concentration of control cells that have been treated the same way except that the media does not contain the candidate drug (but may contain the same level of DMSO). The results of the ELISA are fit to a standard curve and expressed as ng/mL NTP. See W096/40895. <br><br> In a preferred in vitro model system, the AD7c-NTP is cloned into a Lac-Switch inducible system and stably transfected into neuronal cells (e.g., PNET2 (CYZ), SH-Sy5y and hNT2). AD7c-NTP may be the full length cDNA or a CAT reporter gene construct. Protein expression is inducible with 1-5 mM IPTG. Cultures may be examined for cell death, neuritic sprouting and the corresponding changes in gene expression associated with these or other AD-reiated phenomena. Analytical methods available for analysis include, but are <br><br> -24- <br><br> not limited to, viability (Crystal violet) and metabolic (MTT) assays, western blot and immunocytochemical staining, Microtiter ImmunoCytochemical ELISA (MICE) assay, apoptosis DNA fragmentation assays (ladder, end-labeling, Hoechst staining and TUNEL assay) and CAT assay for gene expression studies. <br><br> The effects of candidate drugs on the toxicity of NTP to neuronal cells can also be determined in primary rat cortical cell cultures according to W096/40895, or with human fetal brain tissue, or differentiated neuronal cell lines such as hNT2 and SH-Sy5y cell lines. Alternatively, neuronal cells transformed with and expressing the gene coding for AD7c-NTP as described herein may be used. <br><br> Antisense oligonucleotides have been described as naturally occurring biological inhibitors of gene expression in both prokaryotes (Mizuno et al., Proc. Natl. Acad. Sci. USA 57:1966-1970 (1984)) and eukaryotes (Heywood. Nucleic Acids Res. 14:6771-6772 (1986)), and these sequences presumably function by hybridizing to complementary mRNA sequences, resulting in hybridization arrest of translation (Paterson, et al., Proc. Natl. Acad. Sci. USA, 74:4370-4374 (1987)). <br><br> Antisense oligonucleotides are short synthetic DNA or RNA nucleotide molecules formulated to be complementary to a specific gene or RNA message. Through the binding of these oligomers to a target DNA or mRNA sequence, transcription or translation of the gene can be selectively blocked and the disease process generated by that gene can be halted {see, for example. Jack Cohen. Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press (1989)). The cytoplasmic location of mRNA provides a target considered to be readily accessible to antisense oligodeoxynucleotides entering the cell; hence much of the work in the field has focused on RNA as a target. Currently, the use of antisense oligodeoxynucleotides provides a useful tool for exploring regulation of gene expression in vitro and in tissue culture (Rothenberg, et al.,J. Natl. Cancer Inst. 57:1539-1544(1989)). <br><br> Antisense therapy is the administration of exogenous oligonucleotides which bind to a target polynucleotide located within the cells. For example. <br><br> -25- <br><br> antisense oligonucleotides may be administered systemically for anticancer therapy (WO 90/09180). AD7c-NTP is produced by neuroectodermal tumor cells, <br><br> malignant astrocytoma cells, glioblastoma cells, and in relatively high concentrations (i.e, relative to controls) in brain tissue of AD patients. Thus, <br><br> AD7c-NTP antisense oligonucleotides of the present invention may be active in treatment against AD, as well as neuroectodermal tumors, malignant astrocytomas, and glioblastomas. <br><br> As discussed above, NZ337445 also relates to the correct amino acid and nucleotide sequence for NTP. Thus, NZ337445 also relates to antisense oligonucleotides which are complementary to the mRNA which may be transcribed from Seq. ID No. 1, wherein said oligonucleotides correspond to regions of the NTP gene that were incorrectly sequenced in W094/23756 and W096/15272, e.g. in the region including nucleotides 150-1139 (nucleotides 1-148 of Fig. 16R of published application; nucleotides 1-149 of Seq. ID No. 1 of the present application; were correctly sequenced). This incorrect sequence is present in Seq. ID Nos. 3 and 4. Thus, the invention relates to an antisense oligonucleotide which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No. 1. Preferebly, the oligonucleotides correspond to regions including nucleotides selected from the group consisisting of nucleotides 150, 194-195, 240-241, 243, 244, 255-256, <br><br> 266-267, 269-271, 276, 267, 279-280, 293-295, 338-340, 411, 459, 532-533, <br><br> 591, 633-644, 795-797, 828, 853-854, 876-877, 883, 884-885, 898, 976, 979-980,999,1037,1043-1044,1092-1096,1099, and 1116-1119ofSeq.IDNo. <br><br> 1. More preferably, the invention is related to an antisense oligonucleotide sequence selected from the group consisting of: <br><br> 5' TTC ATC CTG GGT AAG AGT GGG ACA CCT GTG (Seq. ID No. 9); <br><br> 5' TGG TGC ATG TCT TTG GTC CCA GCT AC (Seq. ID No. 10); and 5' ATC AAC CTG GCG AAC ATG GTG AAC CCC ATC (Seq. ID No. 11). <br><br> Also preferably, the sequence is a 15 to 40-mer, more preferably, a 15 to 30-mer. Also preferably, the antisense oligonucleotide it a phosphorothioate or <br><br> Intellectual property' <br><br> office of n.z <br><br> 2 6 AUG 20M <br><br> -26- <br><br> one of the other oligonucleotide derivatives mentioned above. Also preferred are antisense oligonucleotides which are complementary to an NTP nucleic acid sequence and which are nonhomologous to PTP nucleic acid sequences and that correspond to regions that were incorrectly sequenced in the past, as well as 5 pharmaceutical compositions comprising such oligonucleotides and a pharmaceutically acceptable carrier. <br><br> Useful to the present invention are pharmaceutical compositions comprising an effective amount of at least one of the NTP antisense oligonucleotides in combination with a pharmaceutically <br><br> 10 acceptable carrier. In one embodiment, a single NTP antisense oligonucleotide is utilized. In another embodiment, two NTP antisense oligonucleotides are utilized which are complementary to adjacent regions of the NTP DNA. Administration of two NTP antisense oligonucleotides which are complementary to adjacent regions of the DNA or corresponding mRNA may allow for more efficient 15 . inhibition of NTP genomic transcription or mRNA translation, resulting in more effective inhibition of NTP production. <br><br> Preferably, the NTP antisense oligonucleotide is coadministered with an agent which enhances the uptake of the antisense molecule by the cells. For example, the NTP antisense oligonucleotide may be combined with a lipophilic 20 cationic compound which may be in the form of liposomes. The use of liposomes to introduce nucleotides into cells is taught, for example, in U.S. Patent Nos. <br><br> 4,897,355 and 4,394,448. See also U.S. Patent Nos. 4,235,871, 4,231,877, <br><br> 4,224,179, 4,753,788, 4,673,567, 4,247,411, 4,814,270 for general methods of preparing liposomes comprising biological materials. <br><br> 25 Alternatively, the NTP antisense oligonucleotide may be combined with a lipophilic carrier such as any one of a number of sterols including cholesterol, <br><br> cholate and deoxycholic acid. A preferred sterol is cholesterol. <br><br> In addition, the NTP antisense oligonucleotide may be conjugated to a peptide that is ingested by cells. Examples of useful peptides include peptide 30 hormones, antigens or antibodies, and peptide toxins. Rv rhnostntr a npntide &amp; <br><br> ifeECTUALPROPEHTV ofhce of n2 <br><br> 2 6 AUG 200'i <br><br> RECEIVED <br><br> -27- <br><br> is selectively taken up by the neoplastic cells, specific delivery of the antisense agent may be effected. The NTP antisense oligonucleotide may be covalently bound via the 5'OH group by formation of an activated aminoalkyl derivative. The peptide of choice may then be covalently attached to the activated NTP antisense oligonucleotide via an amino and sulfhydryl reactive hetero bifunctional reagent. The latter is bound to a cysteine residue present in the peptide. Upon exposure of cells to the NTP antisense oligonucleotide bound to the peptide, the peptidyl antisense agent is endocytosed and the NTP antisense oligonucleotide binds to the target NTP mRNA to inhibit translation (Haralambidef al., WO 8903849; Lebleu et al, EP 0263740). <br><br> The NTP antisense oligonucleotides described above, and the pharmaceutical compositions of the present invention, may be administered by any means that achieve their intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intra-peritoneal. transdermal, intrathecal or intracranial routes. The dosage administered will be dependent upon the age, <br><br> health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired. <br><br> Useful compositions include all compositions wherein the NTP antisense oligonucleotide is contained in an amount effective to achieve inhibition of proliferation and/or stimulate differentiation of the subject cancer cells, or alleviate AD. While individual needs vary, determination of optimal ranges of effective amounts of each component is with the skill of the art. <br><br> Typically, the NTP antisense oligonucleotide may be administered to mammals, e.g. humans, at a dose of 0.005 to 1 mg/kg/day, or an equivalent amount of the pharmaceutically acceptable salt thereof, per day of the body weight of the mammal being treated. <br><br> Antisense oligonucleotides can be prepared which are designed to interfere with transcription of the NTP gene by binding transcribed regions of duplex DNA (including introns, exons, or both) and forming triple helices (U.S. 5,594,121, U.S. <br><br> 5,591,607, W096/35706, W096/32474, W094/17091, W094/01550, WO <br><br> 41 <br><br> -28- <br><br> intellecfual property office of n.z. <br><br> 2 1 FEB 2002 received <br><br> 91/06626, WO 92/10590). Preferred oligonucleotides for triple helix formation are oligonucleotides which have inverted polarities for at least two regions of the oligonucleotide (Id). Such oligonucleotides comprise tandem sequences of opposite polarity such as 3'—5'-L-5'—3', or 5'—3-L-3'—5', wherein L represents a 0-10 base oligonucleotide linkage between oligonucleotides. The inverted polarity form stabilizes single-stranded oligonucleotides to exonuclease degradation (Froehler et al., supra). Preferred oligonucleotides are nonhomologous to PTP nucleic acid sequences, and correspond to regions that were incorrectly sequenced in the past. The invention is related as well to pharmaceutical compositions comprising such oligodeoxynucleotides and a pharmaceutically acceptable carrier. <br><br> In therapeutic application, the triple helix-forming oligonucleotides can be formulated in pharmaceutical preparations for a variety of modes of administration, including systemic or localized administration, as described above. <br><br> The antisense oligonucleotides and triple helix-forming oligonucleotides may be prepared according to any of the methods that are well known to those of ordinary skill in the art, including methods of solid phase synthesis and other methods as disclosed in the publications, patents and patent applications cited herein. <br><br> NZ337445 is also directed to ribozymes comprising a target sequence which is complementary to an NTP sequence of Seq. ID No. 1 and nonhomologous to PTP nucleic acid sequences and that correspond to regions that were incorrectly sequenced in the past, as well as pharmaceutical compositions comprising such ribozymes and a pharmaceutically acceptable carrier. <br><br> Ribozymes provide an alternative method to inhibit mRNA function. Ribozymes may be RNA enzymes, self-splicing RNAs, and self-cleaving RNAs (Cech et al.. Journal of Biological Chemistry 257:17479-17482 (1992)). It is possible to construct de novo ribozymes which have an endonuclease activity directed in trans to a certain target sequence. Since these ribozymes can act on various sequences, ribozymes can be designed for virtually any RNA substrate. <br><br> -29- <br><br> Thus, ribozymes are very flexible tools for inhibiting the expression of specific genes and provide an alternative to antisense constructs. <br><br> A ribozyme against chloramphenicol acetyltransferase mRNA has been successfully constructed (Haseloffe/ al, Nature 354:585-591 (1988);Uhlenbeck et al. Nature 328:596-600 (1987)). The ribozyme contains three structural domains: 1) a highly conserved region of nucleotides which flank the cleavage site in the 5' direction; 2) the highly conserved sequences contained in naturally occurring cleavage domains of ribozymes, forming a base-paired stem; and 3) the regions which flank the cleavage site on both sides and ensure the exact arrangement of the ribozyme in relation to the cleavage site and the cohesion of the substrate and enzyme. RNA enzymes constructed according to this model have already proved suitable in vitro for the specific cleaving of RNA sequences (Haseloff et al, supra). Examples of such regions include the antisense oligonucleotides mentioned above. <br><br> Alternatively, hairpin ribozymes may be used in which the active site is derived from the minus strand of the satellite RNA of tobacco ring spot virus (Hampel etal, Biochemistry 28:4929-4933 (1989)). Recently, a hairpin ribozyme was designed which cleaves human immunodeficiency virus type 1 RNA(Ojwang et al, Proc. Natl. Acad. Sci. USA 59:10802-10806 (1992)). Other self-cleaving RNA activities are associated with hepatitis delta virus (Kuo et al., J. Virol. (52:4429-4444 (1988)). See also U.S. 5,574,143 for methods of preparing and using ribozymes. Preferably, the NTP ribozyme molecule of the present invention is designed based upon the chloramphenicol acetyltransferase ribozyme or hairpin ribozymes. described above. Alternatively, NTP ribozyme molecules are designed as described by Eckstein et al. (International Publication No. WO 92/07065) who disclose catalytically active ribozyme constructions which have increased stability against chemical and enzymatic degradation, and thus are useful as therapeutic agents. <br><br> In an alternative approach, an external guide sequence (EGS) can be constructed for directing the endogenous ribozyme, RNase P, to intracellular NTP <br><br> intellectual- property office of n.z. <br><br> 2 1 FEB 2002 <br><br> received <br><br> -30- <br><br> mRNA, which is subsequently cleaved by the cellular ribozyme (Altman et al, U.S. Patent No. 5,168,053). Preferably, the NTP EGS comprises a ten to fifteen nucleotide sequence complementary to AD7c-NTP mRNA (corresponding to the miss-sequenced regions) and a 3'-NCCA nucleotide sequence, wherein N is preferably a purine (Id.). After NTP EGS molecules are delivered to cells, as described below, the molecules bind to the targeted NTP mRNA species by forming base pairs between the NTP mRNA and the complementary NTP EGS sequences, thus promoting cleavage of NTP mRNA by RNase P at the nucleotide at the 5'side of the base-paired region (Id.). <br><br> Examples of such external guide sequences are: <br><br> CAC TGC ACT TNC CA (Seq. ID No. 12) <br><br> CCA GGT GTA GNC CA (Seq. ID No. 13) <br><br> CAA GGT CCA GNC CA (Seq. ID No. 14) <br><br> Also useful to the present invention are pharmaceutical compositions comprising an effective amount of at least one NTP antisense oligonucleotide, triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS of the invention in combination with a pharmaceutically acceptable carrier. Preferably, the NTP antisense oligonucleotide, triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS is coadministered with an agent which enhances the uptake of the NTP antisense oligonucleotide, triple helix-forming oligonucleotide, ribozyme or NTP EGS molecule by the cells. For example, the NTP antisense oligonucleotide, triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS may be combined with a lipophilic cationic compound which may be in the form of liposomes, as described above. Alternatively, the NTP antisense oligonucleotide, NTP triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS may be combined with a lipophilic carrier such as any one of a number of sterols including cholesterol, cholate and deoxycholic acid. A preferred sterol is cholesterol. <br><br> The NTP antisense oligonucleotide, NTP triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS, and the pharmaceutical compositions of the present invention may be administered by any means that achieve their <br><br> intellectual property office of n.z. <br><br> - 2 1 FEB 2002 <br><br> received <br><br> -31- <br><br> intended purpose. For example, administration may be by parenteral, subcutaneous, intravenous, intramuscular, intra-peritoneal, transdermal, intrathecal or intracranial routes. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the^ nature of the effect desired. For example, as much as 700 milligrams of antisense oligodeoxynucleotide has been administered intravenously to a patient over a course of 10 days (i.e., 0.05 mg/kg/hour) without signs of toxicity (Sterling, "Systemic Antisense Treatment Reported," Genetic Engineering News 12(12):l, 28 (1992)). <br><br> Useful compositions include all compositions wherein the NTP antisense oligonucleotide, NTP triple helix-forming oligonucleotide, NTP ribozyme or NTP EGS is contained in an amount which is effective to achieve inhibition of proliferation and/or stimulate differentiation of the subject cancer cells, or alleviate AD. While individual needs vary, determination of optimal ranges of effective amounts of each component is with the skill of the art. <br><br> In addition to administering the NTP antisense oligonucleotides, triple helix-forming oligonucleotides, ribo2ymes. or NTP EGS as a raw chemical in solution, the therapeutic molecules may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the NTP antisense oligonucleotide, triple helix-forming oligonucleotide, ribozyme, or NTP EGS into preparations which can be used pharmaceutically. Suitable formulations for parenteral administration include aqueous solutions of the NTP antisense oligonucleotides, NTP triple helix-forming oligonucleotides, NTP ribozymes, NTP EGS in water-soluble form, for example, water-soluble salts. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides. Aqueous injection suspensions may contain substances <br><br> intellectual property office of n.z. <br><br> 2 1 FEB 2002 <br><br> -32- <br><br> received which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers. <br><br> Alternatively, NTP antisense oligonucleotides, NTP triple helix-forming oligonucleotides, NTP ribozymes, and NTP EGS can be coded by DNA constructs which are administered in the form of virions, which are preferably incapable of replicating in vivo (see, for example, Taylor, WO 92/06693). For example, such DNA constructs may be administered using herpes-based viruses (Gage et al., U.S. Patent No. 5.082,670). Alternatively, NTP antisense oligonucleotides, NTP triple helix-forming oligonucleotides, NTP ribozymes, and NTP EGS can be coded by RNA constructs which are administered in the form of virions, such as retroviruses. The preparation of retroviral vectors is well . known in the art (see, for example, Brown et al., "Retroviral Vectors," in DNA Cloning: A Practical Approach, Volume 3, IRL Press, Washington, D.C. (1987)). <br><br> Gene therapy can be used to alleviate AD by inhibiting the inappropriate expression of a particular form of NTP. Moreover, gene therapy can be used to alleviate AD by providing the appropriate expression level of a particular form of NTP. In this case, particular NTP nucleic acid sequences may be coded by DNA or RNA constructs which are administered in the form of viruses, as described above. Alternatively, "donor cells" may be modified in vitro using viral or retroviral vectors containing NTP sequences, or using other well known techniques of introducing foreign DNA into cells (see, for example, Sambrook et al, supra). Such donor cells include fibroblast cells, neuronal cells, glial cells, and connective tissue cells (Gage et al, supra). Following genetic manipulation, the donor cells are grafted into the central nervous system and thus, the genetically-modified cells provide the therapeutic form of NTP (Id.). <br><br> Moreover, such virions may be introduced into the blood stream for delivery to the brain. This is accomplished through the osmotic disruption of the blood brain barrier prior to administration of the virions (see, for example, <br><br> -JJ- <br><br> Neuwelt, United States Patent No. 4,866,042). The blood brain barrier may be disrupted by administration of a pharmaceutically effective, nontoxic hypertonic solution, such as mannitol, arabinose, or glycerol (Id.). <br><br> Having now generally described the invention, the same will be more readily understood through reference to the following Examples which are provided by way of illustration, and are not intended to be limiting of the present invention, unless specified. <br><br> Examples Example 1 Isolation of the AD 7c-NTP cDNA <br><br> A cDNA library was prepared commercially (Invitrogen Corp., San Diego, C A) using RNA extracted from the temporal lobe of an individual with end-stage AD. The library was ligated into the pcDNA2 vector (InVitrogen). To isolate the AD7c-NTP gene, approximately 5x105 transformed and IPTG induced (Sambrook, J. et al. "Molecular Cloning. A Laboratory Manual," Cold Spring Harbor Laboratory Press, Cold Spring Harbor. N.Y.) E. coli colonies were screened using polyclonal antibodies to human PTP (Gross, J. et al, "Isolation, Characterization, and Distribution of an Unusual Pancreatic Human Secretory Protein," J. Clin. Invest. 7(5:2115-2126 (1985)), followed by radiolabeled anti-human IgG (Amersham, Arlington Heights, IL) (Sambrook, J. et al. (1989); and Ausubeh F.M. et al., "Current Protocols in Molecular Biology," New York, NY, John Wiley &amp; Sons (1988)). Restriction endonuclease fragments (Xhol-Pstl; Pstl-Pvull; Pvull-Hindlll) of AD7c-NTP were subcloned into pGem7 (Promega Corp., Madison, WI), and the nucleotide sequence of both strands was determined by the dideoxy chain termination method using T7 DNA polymerase (Ausubel, F.M. et al. (1988)). Additional gene specific primers were generated to generate sequences that overlapped the fragments. The DNA sequence was assembled with <br><br> -34- <br><br> the MacVector Software version 4.5 and analyzed using a Sequence Analysis Software of the Genetics Computer Group version 7.3 as implemented on a MicroVax II computer. Database searches were performed using the BLAST network service of the National Center for Biotechnology Information. <br><br> Characteristics of the AD7c-NTP cDNA Isolated from an AD Brain Library <br><br> The AD7c-NTP cDNA contains 1442 nucleotides and begins with an oiigo-dT track. The nucleotide sequence contains an 1125-nucleotide open reading frame starting with the first AUG cod6n, and a 302-nucleotide untranslated sequence that contains an AATAAA polyadenylation signal (Fig. 1). Bestfit and GAP analysis revealed the presence of four Alu-type sequences embedded in the open reading frame (nucleotides 1-170,423-593,595-765, and 898-1068), and a near-duplication (85% identical) of the first 450 nucleotides starting at nucleotide 898. BLAST database comparisons disclosed 3 regions of significance (67-89%) homology to the Huntington's disease region, chromosome 4pl6.3 (Gusella, J.F. et al., "A Polymorphic DNA Marker Genetically Linked to Huntington's Disease," Nature 50(5:24-238 (1983)) (Figure 1), but no alignment with the IT15 Huntington cDNA which contains longer than normal (CAG)n repeats in individuals with Huntington's disease (The Huntington's Disease Collaborative Research Group," ANovel Gene Containing a Trinucleotide Repeat that is Expanded and Unstable on Huntington's Disease Chromosomes." Cell 72:971-983 (1993)). <br><br> The translated 375 amino acid sequence has a predicted molecular weight of 41,718 and estimated pi of 9.89, and is rich in Ser (11.7%) and Pro (8.8%) residues. Kyte-Doolittle and Chou-Fasman hydrophilicity and Hopp-Woods surface probability profiles predict a 15 amino acid hydrophobic leader sequence, and 7 membrane-spanning regions. Corresponding with the organization of the cDNA. subsequent analysis of the protein revealed four 83% to 91% identical repeated (once or twice) antigenic domains between 9 and 23 amino acids in <br><br> length (Fig. 1). Protein subsequent analysis demonstrated 21 cAMP, calmodulin-dependent protein kinase II, protein kinase C, or glycogen synthase kinase 3 phosphorylation sites, and one myristyration site. In addition, a single tgf motif (Residues #44-#53) was detected. Comparison of the AD7c-NTP amino acid sequence with the genebank database revealed four regions of significant homology to the 0-subunit of integrin (72%-80%), the alternatively spliced A4 form of the neurofibromatosis 2 gene (72%-81%), myelin oligodendroglial glycoprotein-16 precursor protein (70%-76%), human integral membrane protein (55%-85%), and human decay accelerating factor 2 precursor (62%-68%), and two regions with homology to the c-rel protooncogene transforming protein (Residues 56-84: 65%; Residues 287-295: 88%) (Fig. 1). Residues 5-24 are 75% identical to a region of the IGF 1 /insulin receptor hybrid, and residues 4-24,47-79, 109-132, 227-261, and 227-360 exhibit 57% to 76% identity with the human transformation-related protein. In addition, two serine/threonine kinase protein domains (Residues 6-48, and 272-294) were identified. <br><br> The in vitro translated protein and pTrcHis-AD7c-NTP recombinant protein purified by metal chelate chromatography and cleaved from the fusion partner had molecular masses of -39-42 kD by SDS-PAGE or Western blot analysis (Fig. 2). In addition, in Bosc cells transfected with the AD7c-NTP cDNA ligated into the pcDNA3 vector (Invitrogen, San Diego, CA), a single -39-42 kD protein was detected by Western blot analysis using the N3I4 monoclonal antibody. In two-site immunoradiometric assays and immunoblotting studies, the AD7c-NTP recombinant protein exhibited specific immunoreactive binding with all of the polyclonal and monoclonal antibodies generated with purified pTrcHis-AD7c-NTP recombinant protein. No immunoreactivity with AD7c-NTP was detected using pre-immune rabbit sera, non-relevant rabbit polyclonal antibodies to GAP-43, or non-relevant monoclonal antibodies to Dengue virus or FB50 (Fig. 2). <br><br> Example 2 In Vitro Expression of AD7c-NTP <br><br> Antisense and sense cRNAs were transcribed from AD7c-NTP cDNA plasmid with Kpnl andXhol, respectively. The cRNA transcripts were translated in a rabbit reticulocyte lysate system (Stratagene, La Jolla, CA) in the presence of [35S]methionine (Dupont-New England Nuclear, Boston, MA), and the products of in vitro translation were analyzed by SDS-PAGE and autoradiography. The AD7c-NTP cDNA was ligated into the pTrcHis expression vector (Invitrogen Corp., San Diego, CA) which encodes a 5 N 6-His Tag sequence used to isolate the fusion protein by metal chelate chromatography. Recombinant fusion protein induction in transformed E. coli was achieved by the addition of 1 mM IPTG during log phase growth. The fusion protein was affinity purified (Ausubel, F.M. et al. (1988)) using ProBond resin (Invitrogen Corp., San Diego, CA), and detected by Western blot analysis (Harlow, E. and Lane, D.. "Antibodies: A Laboratory Manual," Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1988)) with antibodies to do T7-tag fusion partner (Novogen). The tag was then cleaved with entrokinase to give the AD7c-NTP protein. (Ausubel, R.M et al. (eds.) in Current Protocols in Molecular Biology, John Wiley &amp; Sons. Inc., New York, N.Y., 1994. <br><br> Example 3 Generation of Polyclonal and Monoclonal Antibodies to Recombinant AD7c-NTP <br><br> Polyclonal antibodies were generated in rabbits immunized with affinity purified recombinant AD7c-NTP protein. Monoclonal antibodies were generated in Balb/c mice immunized intraperitoneally with 50 ng of purified recombinant AD7c-NTP protein emulsified in complete Freund's adjuvant (Harlow, E. and Lane, D. (1988); and Wands, J.R. and Zurawski, V.R., Jr., "High Affinity Monoclonal Antibodies to Hepatitis B surface Antigen (HBsAg) produced by Somatic Cell Hybrids," Gastroeneology 801:225-232 (1981)). The mice were boosted 6 to 10 weeks later, with 10 fig AD7c-NTP by tail vein injection. <br><br> -37- <br><br> Spleenocytes were fused with SP-0 myeloma cells (Harlow, E. and Lane, D. (1988) and Wands, J.R. and Zurawski, V.R., Jr. (1981)). The cells were grown in HAT medium, and hybridomas producing anti-AD7c-NTP antibody were identified by solid phase immunoassay (Bellet, D.H. etal., "Sensitive and Specific 5 Assay for Human Chorionic Gonadotropin Based on Anti-Peptide and <br><br> Anti-Glycoprotein Monoclonal Antibodies: Construction and Clinical Implications," J. Clin. Endocrinol. Metabol. (53:1319-1327 (1988)). The binding specificity of the immunoglobulin fractions of polyclonal immune sera and the hvbridoma supematants was confirmed by radioimmunoassay (RIA) and Western 10 blot analysis with recombinant AD7c-NTP, and by Western blot analysis and immunohistochemical staining of AD and aged control brains. In a panel of 25 hybridomas, 3 MoAbs exhibited similar levels of immunoreactivity with purified native PTP and recombinant AD7c-NTP, and therefore were further characterized (Table 1). <br><br> 5 <br><br> 10 <br><br> 15 <br><br> 20 <br><br> Table 1 <br><br> Profiles of Immunoreactivity Exhibited by AD7c-NTP Monoclonal Antibodies <br><br> Antibody <br><br> Western Blot <br><br> ICC* <br><br> AD Specific** <br><br> Distribution of Labeling in AD Brains <br><br> N2B10 <br><br> Y: Reducing <br><br> Negative <br><br> N/A <br><br> None <br><br> N2I5 <br><br> Y: Reducing <br><br> Negative <br><br> N/A <br><br> None <br><br> N2J1 <br><br> Yes <br><br> ++ <br><br> Yes <br><br> Neuropil threads, irregular neurites, axons <br><br> N2R1 <br><br> No <br><br> Negative <br><br> N/A <br><br> None <br><br> N2S6 <br><br> Y: Reducing <br><br> ++++ <br><br> Yes <br><br> Neurons <br><br> N2T8 <br><br> Y: Reducing <br><br> ++++ <br><br> Yes <br><br> Degenerating neurons, NFT, irregular neurites <br><br> N2U6 <br><br> Yes <br><br> +++ <br><br> Yes <br><br> Neuropil threads, NFT <br><br> N3A13 <br><br> No <br><br> + <br><br> No <br><br> None <br><br> N3CI1 <br><br> No <br><br> ++ <br><br> No <br><br> None <br><br> N3D12 <br><br> No <br><br> + <br><br> No <br><br> None <br><br> N3I4 <br><br> Y: Nonreducing <br><br> Negative <br><br> N/A <br><br> None <br><br> N2-36 <br><br> No <br><br> + <br><br> Yes <br><br> NFT, Swollen neurites <br><br> N2-22-11 <br><br> No <br><br> Negative <br><br> N/A <br><br> None <br><br> Polyclonal <br><br> Yes <br><br> Yesf <br><br> Degenerating neurons, irregular neurites <br><br> *ICC = Immunocytochemistry; NFT = neurofibrillary tangles <br><br> **AD-specific: Immunoreactivity only detected in histologically normal neutrons and fibers in AD tissue sections <br><br> (N2S6), or in degenerating neuronal cell bodies and processes detected in AD. <br><br> t = following formic acid treatment only. <br><br> co <br><br> 00 <br><br> 1 <br><br> -39- <br><br> Profiles ofAD7c-NTP Immunoreactivity Revealed with MoAbs (Table J): The findings summarized below are representative of the observations made in 6 end-stage AD and 5 aged control brains. Twenty-five of the AD7c-NTP MoAbs were characterized by immunocytochemical staining. Table 1 details features of 13 AD7c-NTP MoAbs. The other 12 MoAbs were excluded from the list because either they were not suitable for immunocytochemical staining and Western immunoblot studies due to low-level binding (N=9), or they exhibited cross-immunoreactivity with pancreatic thread protein (N=3). Among the 13 AD7c-NTP MoAbs that were further characterized, only 8 exhibited immunoreactivity in neuronal perikarya, neuropil fibers, white matter fibers (axons), or AD neurodegenerative lesions. The other 5 were non-immunoreactive in histologic sections. <br><br> In Table 1, AD-specific binding refers to the detection of degenerating neurons, neurofibrillary tangles, irregular swollen neurites and axons, or immunoreactivity in histologically intact neurons in AD but not control brains. Four AD7c-NTP MoAbs (N2-36, N3-C11, N2S6, N2-T8) exhibited intense degrees of immunocytochemical staining in cortical neurons, particularly pyramidal cells in layers 3 and 5. Two MoAbs (N2-U6,N2-S6) prominently labeled neuropil and white matter fibers (axons), and 5 (N2-U6, N3-C11, N2-S6, N2-T8. N2-J1) detected A2B5+ and GFAP+ protoplasmic (Type 2) astrocytes in the cerebral cortex and white matter. Two AD7c-NTP MoAbs (N2-U6 and N2-T8) exhibited intense labeling of cortical neurons and swollen, irregular (dystrophic) neuropil neurites in AD, but low-level or absent labeling in aged control brains. Most striking was the immunoreactivity observed in AD-associated neurodegenerative lesions using the N2-36, N2-T8, N2-U6 MoAbs. N2-T8 detected intracellular neurofibrillary tangles as well as degenerated neurons without neurofibrillary tangles; N3-D12, N2-T8, and N2-J1 labeled swollen dystrophic axons and fine neuritic processes, particularly in superficial layers of the cerebral cortex; and N2-U6 and N2-J1 labeled wavy irregular threadlike structures detected only in AD brains. N2J1 very prominently labeled irregular threadlike structures, dystrophic neurites, and swollen axons, but exhibited minimal labeling of neuronal perikarya <br><br> -40- <br><br> or glial cells. The negative control 5C3 MoAb to Hepatitis B vims was not immunoreactive with adjacent sections of the same brains. <br><br> In the Examples which follow, polyclonal and the N3I4, N2J1, and N2U6 monoclonal AD7c-NTP antibodies were employed. <br><br> Example 4 Human Brain Tissue <br><br> Human brain tissue was obtained from the Alzheimer's Disease Research Center brain bank at the Massachusetts General Hospital (MGH-ADRC). All brains were harvested within 12 hr of death, and the histopathological diagnosis of AD was rendered using CERAD criteria (Mirra. S.S. et al., "The Consortium to Establish a Registry for Alzheimer's Disease (CERAD). II. Standardization of the Neuropathological assessment of Alzheimer's Disease," Neurology 47:479-486 (1991)). The AD group (N=17) had a mean age of 76.3±8.8 years, a mean brain weight of 1117±101 grams, and a mean postmortem interval of 7.3±3.9 hours. The control group (N=l 1) had a mean age of 78.0±6.2 years, a mean brain weight of 1274±115 grams, and a mean postmortem interval of 8.3±3.6 hours. In addition, 4 cases of early probable AD with cognitive decline and moderate AD histopathological lesions, and 2 cases of diffuse Lewy body disease (Kosaka. K. "Dementia and Neuropathology in Lewy Body Disease," Adv. Neurol. 60:456-463 (1993)) (DLBD: an AD-related CNS neurodegenerative disease) were studied. Fresh frozen frontal and temporal lobe tissue was used for Northern and Western blot analyses. Post-mortem cerebrospinal fluid (CSF) samples (8 AD; 7 control; 2 DLBD) were used to detect AD7c-NTP by Western blot analysis. Paraffin-embedded histological sections were used to localize AD7c-NTP gene expression by in situ hybridization and immunohistochemical staining. <br><br> -41 - <br><br> Examples Northern Analysis of AD7c-NTPmRNA Expression <br><br> Samples (15 jig) of total RNA isolated (Ausubel, F.M. et al. (1988)) from AD and aged control frontal lobe tissue (Brodmann Area 11), and normal adult human kidney, liver, spleen, gastrointestinal tract, ovaries, fallopian tubes, uterus, thyroid, lung, skeletal muscle, and pancreas, were subjected to Northern hybridization analysis using 2xl06 dpm/ml of [a32P]dCTP-labeled AD7c-NTP cDNA probe (specific activity ~108 dpm/jjg DNA) generated by the random hexamer method (Ausubel, F.M. et al. (1988)). The blots were subsequently washed in stepwise dilutions of 5x SSC (IX SSC is 0.15 M NaCl plus 0.015 M sodium citrate) containing 0.5% SDS (Sambrook, J. et al. (1989); Ausubel, F.M. et al. (1988)), and finally in O.lx SSC/0.5% SDS at 65°C. To evaluate RNA loading, the blots were stripped of probe and re-hybridized with a 10-fold molar excess of a [y^PjATP-labeled synthetic 30mer corresponding to 18s ribosomal RNA (de la Monte, S.M. and Bloch, K.D. (1996)). The results were analyzed by autoradiography and densitometry (ImageQuant, Molecular Dynamics. Inc). <br><br> Results: AD 7c-NTP mRNA Expression in AD and Aged Control Brains <br><br> In Northern blot hybridization studies. AD7c-NTP cDNA probes detected 1.4 kB and 0.9 kB mRNA transcripts in adult human frontal and temporal lobe tissue, but not pancreas, kidney, liver, spleen, gastrointestinal tract (various regions) ovaries, fallopian tubes, uterus, thyroid, lung, skeletal muscle, testis, and thymus were negative. Both 1.4 kB and 0.9 kB AD7c-NTP mRNA transcripts were detected in AD and aged control brains, but the levels of expression were increased in AD. With values normalized to 18S RNA signals to correct for differences in loading and non-specific degradation, densitometric analysis of non-saturated autoradiograms revealed significantly higher mean levels of both the 1.4 kB (P&lt;0.01)and the 0.9 kB (P&lt;0.05) AD7c-NTP transcripts in AD compared with normal aged control brains. <br><br> -42- <br><br> Example 6 Reverse Transcriptase-polymerase Chain Reaction Amplification (RT-PCR) Studies <br><br> Samples of total RNA (2 (j.g) isolated from human brain, PNETl and PNET2 human CNS neuronal cell lines (The, I. et al., "Neurofibromatosis type 1 Gene Mutations in Neuroblastoma," Nature Genet. 3:62-66 (1993)) (positive controls), SH-Sy5y human neuroblastoma cells (Biedler, J.L.et al., "Morphology and Growth, Tumorigenicity, and Cytogenetics of Human Neuroblastoma Cells in Continuous Culture," Cancer Res. 33:2643-2652 (1973)), and human pancreas and liver (negative controls) were reverse transcribed using random hexamer primers (Ausubel, F.M. (1988)) and Superscript™ reverse transcriptase (Gibco-BRL, Grand Island, NY). The cDNA products (10%) were subjected to PCR amplification to detect AD7c-NTP sequences using the primers: (459-480) 5' TGTCCCACTCTTACCCAGGATG [Seq ID No. 5] and (849-826) 5' AAGCAGGCAGATCACAAGGTCCAG [Seq. ID No. 6]. p-actin control primers (Dallman, M.J. and Porter, A.C.G., "Semi-Quantitative PCR for the Analysis of Gene Expression," In: PCR A Practical Approach, M.J. McPherson et al. (eds.), IRL Oxford University Press, Oxford, pp. 215-224 (1991)) (5' AATGG ATG ACG ATATCGCTG [Seq. ID No. 7]; 5'-ATGAGGTAGTCTGTCAGGT [Seq. ID No. 8]) were incorporated into all studies. Each cycle of PCR amplification consisted of denaturation at 95 °C for 30 sees, annealing at 60°C for 30 sees, and extension at 72 °C for 1 min. After 30 cycles and a final 10 minute extension at 72 °C, approximately 10 percent of the PCR products were analyzed by agarose gel electrophoresis and Southern hybridization using a [y32P]dATP-labeled oligonucleotide probe corresponding to nucleotides 702-720 of the AD7c-NTP cDNA. The remaining PCR products were electrophoretically fractionated and Iigated into PCRII TA cloning vectors (InVitrogen Corp, San Diego, CA). The nucleotide sequences of clones isolated from 6 brain samples were determined by the dideoxy chain termination method (Sambrook, J. et al., (1989); Ausubel, F.M. (1988)). <br><br> Expression of AD7c-NTP mRNA in human brain was verified by RT-PCR amplification of RNA isolated from 6 AD and 5 aged control brains (frontal lobe). <br><br> -43 - <br><br> The expected 390 nucleotide PCR product was obtained with all samples. The specificity of the PCR products was demonstrated by Southern blot analysis using [32P]-labeled oligonucleotide probes corresponding to internal sequences, and by determining that the nucleic acid sequences of the 390-nucleotide PCR products cloned from 5 AD brains were identical to the sequence underlined in Fig. 1. In the RT-PCR amplification studies, AD7c-NTP PCR products were also detected using RNA isolated from PNETl, PNET2 and SH-Sy5y neuronal cells, but not human pancreas or liver. All samples analyzed yielded positive RT-PCR products using the P-actin primers. <br><br> Example 7 In Situ Hybridization <br><br> Paraffin sections (10 fim thick) of AD and control brains were hybridized with antisense and sense (negative control) AD7c-NTP cRNA probes (de la Monte S.M. et al, (1995); de la Monte, S.M. and Bloch, K.D. (1996)) generated from cDNA templates linearized with Kpnl or Xhol, and labeled with [11 -digoxigeninjUTP using SP6 to T7 DNA dependent RNA polymerase (Melton, D.A. et al. "Efficient in Vitro Synthesis of Biologically Active RNA and RNA Hybridization Probes from Plasmids Containing a Bacteriophage SP6 Promoter," Nucl. Acids Res. 72:7035-7056 (1984)). Specifically bound probe was detected with alkaline phosphatase-conjugated sheep F(ab'), anti-digoxigenin (Boehringer-Mannheim Inc.) and X-phosphate/5-bromo-4-chloro-3-indolyl-phosphate/nitro-blue-tetrazolium-chl oride (de la Monte, S.M. and Bloch, K.D. (1996)). The probe specificity was confirmed by Northern blot analysis of brain using identical cRNA probes labeled with [cc32P]UTP. <br><br> Results: Cellular Localization of AD7c-NTP mRNA in Human Brain <br><br> In situ hybridization studies using [ 11 -digoxigenin]UTP-labeled antisense cRNA probes demonstrated AD7c-NTP-related mRNA transcripts in frontal <br><br> -44- <br><br> (Brodmann Area 11) and temporal (Brodmann Area 21) cortex neurons in both AD (N=6) and aged control (N=4) brains (Fig. 3). However, darkfleld microscopy revealed strikingly elevated levels of AD7c-NTP mRNA expression in both temporal and frontal cortex neurons in AD relative to aged control brains, corresponding with the results of Northern blot analysis. Low levels of AD7c-NTP mRNA transcripts were also detected in cortical and white matter glial cells in AD. AD7c-NTP mRNA transcripts were not detected in cerebral blood vessels, and specific hybridization signals were not observed in any of the specimens hybridized with digoxigenin-labeled sense strand cRNA probes. <br><br> Example 7 Immunodetection of AD7c-NTPExpression <br><br> Western immunoblotting studies (Harlow, E. and Lane, D. (1988)) were performed using protein extracts (60 pg samples) generated from postmortem frontal and temporal lobe tissue, and various non-CNS tissues homogenized in RIPA buffer (Ausubel, F.M. et al. (1988)). In addition, 40 jj.1 samples of postmortem or antemortem cerebrospinal fluid were evaluated by Western blot analysis. The blots were probed with rabbit polyclonal (1:800) or N3I4, N2U6, or N2J1 mouse monoclonal (5 pg/ml) anti-AD7c-NTP. Antibody binding was detected with horseradish peroxidase-conjugated secondary antibody diluted 1:25,000 (Pierce), and Supersignal enhanced chemiluminescence reagents (Pierce). The levels of AD7c-NTP expression were quantified by volume densitometric scanning of the autoradiograms (ImageQuant; Molecular Dynamics Inc., Sunnyvale, CA). Cellular localization of AD7c-NTP immunoreactivity was demonstrated in paraffin-embedded histological sections of frontal (Brodmann Area 11) and temporal (Brodmann Area 21) lobe from AD and age-matched control brains. The sections were immunostained by the avidin-biotin horseradish peroxidase complex method (de la Monte, S.M. et al. (1995); and de la Monte, S.M. and Bloch, K.D. (1996)) using the N2J1 and N2U6 AD7c-NTP monoclonal antibodies. Adjacent sections were immunostained with monoclonal antibodies <br><br> -45 - <br><br> to glial fibrillary acidic protein as a positive control, and with monoclonal antibodies to Dengue virus as a negative control. <br><br> Results: Characterization of AD7c-NTP Antibody Binding by Western Blot Analysis <br><br> In Western immunoblotting studies of protein extracted from human frontal and temporal lobe tissue, broad -39-45 kD bands of AD7c-NTP immunoreactivity were detected with the polyclonal and 11 of the 25 monoclonal antibodies. When the proteins were electrophoretically fractionated in 15% Laemmli gels and probed with the N3I4. N2U6. or N2J1 monoclonal antibodies, the -39-45 kD AD7c-NTP-immunoreactive molecules were resolved into 3 or 4 tightly clustered bands (Fig. 2E), possibly representing different degrees of AD7c-NTP phosphorylation. In addition, the polyclonal and 4 of the monoclonal antibodies detected 18-21 kD AD7c-NTP-immunoreactive proteins in brain (Fig. 2E). Western blot analysis of non-CNS tissues revealed no specific binding with the AD7c-NTP antibodies. <br><br> Example 8 In vitro Expression Studies <br><br> The AD7c-NTP cDNA was ligated into the pcDNA3 mammalian expression vector which contains a CMV promoter (InVitrogen, San Diego, CA). SH-Sy5y cells were transfected with either pcDNA3-AD7c-NTP or pcDNA3 (empty vector, negative control), and selected with G418. Stably transfected cell lines were examiner for growth properties, morphology, and expression of AD7c-NTP. Cell growth was assessed by measuring [3H] thymidine incorporation into DNA and determining the density of viable cells in the cultures. Cells grown in chamberslides were immunostained using N3I4 monoclonal antibody. AD7c-NTP expression was also evaluated by Western blot analysis with the N3I4 antibody. <br><br> -46- <br><br> Results: Over-Expression of AD7c-NTP in Neuronal Cells Leads to Apoptosis, Neuritic Sprouting Which are Characteristic of Alzheimer's Disease <br><br> Over-expression of AD7c-NTP in SH-Sy5y neuronal ceils stably transfected with pcDNA3-AD7c-NTP resulted in significantly lower densities of viable cells in the cultures, despite normal or elevated levels of DNA synthesis (Fig. 5). This result was reproducible in other neuronal cells lines and using other expression vectors. Reduced cell density in the cultures was caused by increased cell death. The attendant increase in nuclear p53 expression in AD7c-NTP transfected cells suggests that the cell death is likely to be mediated by apoptosis. Subconfluent cultures of SH-Sy5y cells transfected with pcDNA3 contained round or spindled shaped cells with few or no processes (Fig. 6 A). In contrast, SH-Sy 5y cells transfected with pcDNA3-AD7c-NTP exhibited extensive neuritic growth with fine interconnecting processes detected on most cells (Figs. 6B-6D). In addition, pcDNA3-AD7c-NTP transfected cultures always contained numerous round, refractile floating cells (dead) which failed to exclude Trypan blue dye. Immunocytochemical staining of stationary cultures using the N3I4 monoclonal antibody revealed intense labeling of the cell bodies and cell processes of SH-Sy5y cells transfected with pcDNA3-AD7c-NTP (Figs. 6F and 6G), and absent immunoreactivity in SH-Sy5y cells transfected with pcDNA3 (empty vector) (Fig. 6E). These studies demonstrate that over expression of AD7c-NTP in transfected neuronal cells promotes neuritic sprouting and cell death, two of the major features of Alzheimer's disease neurodegeneration. Thus, transfected cell lines and transgenic animals which over-express the AD7c-NTP will be useful for screening drugs that might be effective in reducing AD7c-NTP expression and, thereby, treating or preventing the onset of Alzheimer's disease. <br><br> -47- <br><br> Example 9 AD7c-NTP Protein Expression in AD and Aged Control Brains <br><br> Western blot analysis and immunohistochemical staining of AD and aged control brains were performed using the N3I4, N2U6, and N2J1 monoclonal AD7c-NTP antibodies. In AD and aged control brains, ~39-45 kD proteins were detected with all 3 monoclonal antibodies. In addition, —18-21 kD proteins were detected with the N2U6 and N2J1 antibodies. Densitometric analysis of the autoradiograms demonstrated significantly higher levels of the -39-45 kD AD7c-NTP in AD relative to aged control frontal lobe tissue (Fig. 2D). In addition, expression of the ~18-21 kD AD7c-NTP-immunoreactive proteins was also increased in AD, but studies have not yet determined whether these molecules represent cleavage products of the -39-45 kD AD7c-NTP, or a unique protein encoded by another cDNA. In a small series, comparisons between early and late AD revealed higher levels of AD7c-NTP immunoreactivity in brains with end-stage disease (Fig. 2E). Using the N3I4 antibody, Western blot analysis detected the presence of -39-45 kD AD7c-NTP molecules in postmortem CSF, and higher levels in AD relative to aged control samples (Fig. 2F). Immunohistochemical staining studies with polyclonal and several brain-specific monoclonal antibodies localized AD7c-NTP immunoreactivity in neurons, neuropil fibers, and white matter fibers in AD and control brains. In immunohistochemical staining studies, the N2U6 and N2J1 antibodies exhibited intense immunoreactivity in intact as well as degenerating cortical neurons and dystrophic neurites in AD brains, but low-level or absent immunoreactivity in aged control brains (Fig. 4). Omission or pre-adsorption of the primary antibody with recombinant AD7c-NTP protein, or the application of non-relevant primary antibody (negative controls) also yielded negative immunostaining results. All sections of brain exhibited positive immunoreactivity with monoclonal antibodies to glial fibrillary acidic protein (positive control). <br><br> These studies demonstrate elevated levels of AD7c-NTP expression in AD relative to aged control brains, and abnormal AD7c-NTP gene expression localized in AD brain neurons by in situ hybridization and immunohistochemical <br><br> -48 - <br><br> staining. Although two distinct mRNA transcripts and at least two distinct protein species were detected in brain, the levels of the mRNA and protein corresponding to AD7c-NTP were increased in AD. We have not yet determined whether the smaller transcripts and protein species are distinct, or represent alternately spliced forms of a single gene. Although the cDNA was isolated from a library prepared with RNA isolated from a single AD brain, the RT-PCR studies confirmed the presence of identical sequences in 6 different AD brains. Since the AD7c-NTP cDNA exhibits no significant primary sequence homology with the human pancreatic protein (Watanabe, T. et al., "Complete Nucleotide Repeat that is expanded and Unstable on Huntington's Disease Chromosomes," Cell 72:971-983 (1993)), the cross-reactivity of polyclonal antibodies with AD7c-NTP molecule probably occurs through conformational epitopes. Increased expression of AD7c-NTP was observed in both histologically intact and degenerating neurons and cell processes, and a recent study suggested that AD7c-NTP protein expression occurs early in AD neurodegeneration. <br><br> Example 10 In Vitro Drug Screening System <br><br> AD7c-NTP was cloned into a Lac-Switch expression vector (Stratgene), and CYZ neuronal cells were stably transformed with the construct. Several cell lines were selected that expressed AD7c-NTP at various levels, Lac A-Lac F, after induction of protein expression with IPTG. Experiments were done to determine the effect ( e.g., change in morphology, gene expression, viability, etc.) of AD7c-NTP expression on neuronal cells, thereby generating markers useful for screening, in vitro, potential pharmacologic agents for the treatment of AD. <br><br> Expression levels were determined by Microtiter ImmunCytochemical Elisa Assay (MICE). Briefly, 104 cells/well were seeded into 96-well plates, and were induced to express AD7c-NTP for a period of 6-18 hrs; additionally, in some experiments, cells were exposed to toxins or protective agents for a similar period of time. At the end the treatment period, cells were fixed, permeabilized and immunostained with the appropriate antibody following the ABS procedure. <br><br> -49- <br><br> Quantitation was done by incubating cells with a soluble chromagen, stopping the reaction with 2M H2S04 and determining chromagen absorbance in an automated ELISA reading machine. After staining with Coommassie Blue, the ratio of immunoreactivity (i.e. bound chromagen) to Coommassie Blue absorbance was determined (MICE units), and the results were graphed. Alternatively, immunoreactivity may also be determined with a precipitating chromagen (e.g. DAB, TruBlue or AEC). <br><br> For Experiments determining cell viability, after culture and treatment as for the MICE assay, culture media was replaced with a Crystal Violet/PBS/formalin solution. After staining, cells were rinsed thoroughly and lysed with a PBS/1% SDS solution, and absorbance was determined with an automated ELSA reader. Results were graphed as percent viability. <br><br> Results: Over-Expression of AD7c-NTP in Neuronal Cells Leads to Alterations in Gene Expression, Cell Viability and Toxin Hypersensitivity <br><br> Expression of AD7c-NTP results in altered expression of genes associated with AD (Tau, bA4 amyloid), neuritic sprouting (synaptophysin) and apoptosis (p53, SC95-Fas, NO-Tyr, NOS3) (Figures 7A-7C and 8A-8D). In Figures 7A-7C, the percent change in expression. 24 hrs. after AD7c-NTP induction, is presented for the indicated genes. In the absence of AD7c-NTP expression (Fig. 7A), little or no change in gene expression is observed in Lac A-control, nonexpressing cells; however, similar experiments done with Lac B (Fig. 7B) and Lac F (Fig.7C) cells induced to express different levels of AD7c-NTP (B6) demonstrate marked changes in gene expression. For example, NOS3 is expressed at almost twice the normal level in the Lac B-B6 experiment (Fig. 7B) than in Lac A-control cells (Fig. 7A). <br><br> Figures 8A-8D demonstrate that altered gene expression is dependent on the level of AD7c-NTP induction. Results are presented for the NTP (Fig. 8A), Synaptohysin (Fig. 8B), Tau (Fig. 8C) and p53 (Fig. 8D) genes as percent change in expression as a function of IPTG induction of AD7c-NTP expression in stably <br><br> -50- <br><br> transfected, CYZ neuronal cells. LacB (filled square) and LacF (open circle) cells were exposed to the indicated amounts of IPTG (1-5 mM) for 24 hrs.. These data indicate that increased concentrations of IPTG leads to an up-regulation of all genes examined. <br><br> Two assays were used to evaluate the effects of AD7c-NTP expression in CYZ neuronal cells: metabolic activity was measured by the MTT assay (Fig. 9A), and cell death was measured by the CV viability assay (Fig.9B). Results are expressed as percent change in MTT activity or cell viability 24 hrs. after IPTG induction relative to untreated, parallel control cultures. Six different clones, LacA-LacF. were assayed after IPTG induction (B6) and compared to LacA control cells lacking AD7c-NTP. For all 6 clones examined, stimulation of AD7c-NTP expression results in substantially reduced metabolic activity relative to control cells (Fig. 9A). Cell death was induced in LacB-B6 and LacF cells, and reduced cell viability was observed in LacA-B6 and LacE-B6 clones. <br><br> Decreased cell viability of cells expressing AD7c-NTP is exacerbated by oxidative stress. When LacB and LacF cells were induced with 3mM IPTG (B6) for 24 hrs or 48 hrs. and exposed to toxins that increase oxidative stress for 6 hrs or 24 hrs. (Figs. 10A and 10B, respectively), cell viability decreased markedly as compared to LacA-control, nonexpressing cells. Results depicted in Figures 10A and 10B establish both that longer AD7c-NTP induced expression and longer exposure to hydrogen peroxide (H202) and diethyldithiocarbamic acid (DDC) lead to decreased cell viability and increased hypersensitivity, respectively. <br><br> In order to determine the reason for decreased cell viability, experiments were done to quantitatively measure apoptosis in stably transfected, CYZ neuronal cells expressing AD7c-NTP; results are presented in Figure 11. The degree of apoptosis was determined by incubating cells in the presence of 32dCTP to measure template-independent incorporation of label into fragmented DNA, a characteristic of the apoptotic mechanism of ceil death. Comparison of control (uninduced, transfected cells) with 3mM induced (indicated by B6) LacA, LacB and LacF cells clearly indicates that expression of AD7c-NTP leads to increased incorporation of 32dCTP label into cellular DNA (increased apoptosis). Variations <br><br> -51 - <br><br> in the incorporation of label between IPTG induced cell lines are attributed to differences in the level of AD7c-NTP expression. <br><br> The established in vitro system provides a means for screening pharmacologic agents that modulate or counteract the changes effected through AD7c-NTP expression and, ostensibly, the AD process. AD7c-NTP expression leads to up-regulation of nitric oxide synthase which, in some neuronal cells, causes oxygen free radical formation. The experiment depicted in Figure 12 establishes that AD7c-NTP induced oxidative stress can be counteracted by pharmacologic agents. Results are expressed as the ratio of percent change in viability for experimental (AD7c-NTP induced) over control, uninduced cells. In Figure 12, CYZ cells, stably transfected with AD7c-NTP, are induced to express AD7c-NTP and exposed to various pharmacologic agents. Hydrogen peroxide (H:02) and diethyldithiocarbamic acid (DDC) exacerbate cell death, while agents such pyroglutamate (PG) (and L-NAME and L-arginine) inhibit or reduce the nitric oxide synthase toxicity attributable to AD7c-NTP expression. <br><br> From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions without undue experimentation. All patents, patent applications and publications cited herein are incorporated by reference in their entirety. <br><br> -52- <br><br> SEQUENCE LISTING <br><br> (1) GENERAL INFORMATION: <br><br> (i) APPLICANT: THE GENERAL HOSPITAL CORPORATION FRUIT STREET ' BOSTON, MA 02114 UNITED STATES OF AMERICA <br><br> APPLICANT/INVENTOR: de la Monte, Suzanne <br><br> Wands, Jack R. <br><br> (ii) TITLE OF INVENTION: Transgenic Animals and Cell Lines for <br><br> Screening Drugs Effective for the Treatment or Prevention of Alzheimer's Disease <br><br> (iii) NUMBER OF SEQUENCES: 14 <br><br> (iv) CORRESPONDENCE ADDRESS: <br><br> (A) ADDRESSEE: Sterne, Kessler, Goldstein &amp; Fox P.L.L.C. <br><br> (B) STREET: 1100 New York Ave., Suite 600 <br><br> (C) CITY: Washington <br><br> (D) STATE: DC <br><br> (E) COUNTRY: USA <br><br> (F) ZIP: 20005-3934 <br><br> (v) COMPUTER READABLE FORM: <br><br> (A) MEDIUM TYPE: Floppy disk <br><br> (B) COMPUTER: IBM PC compatible <br><br> (C) OPERATING SYSTEM: PC-DOS/MS-DOS <br><br> (D) SOFTWARE: Patentln Release #1.0, Version #1.30 <br><br> (vi) CURRENT APPLICATION DATA: <br><br> (A) APPLICATION NUMBER: To be assigned <br><br> (B) FILING DATE: Herewith <br><br> (C) CLASSIFICATION: <br><br> (vii) PRIOR APPLICATION DATA: <br><br> (A) APPLICATION NUMBER: US 60/038,908 <br><br> (B) FILING DATE: 26-FEB-1997 <br><br> (viii) ATTORNEY/AGENT INFORMATION: <br><br> (A) NAME: Esmond, Robert W. <br><br> (B) REGISTRATION NUMBER: 32,893 <br><br> (C) REFERENCE/DOCKET NUMBER: 0609.4 37PC01 <br><br> (ix) TELECOMMUNICATION INFORMATION: <br><br> (A) TELEPHONE: 202-371-2600 <br><br> (B) TELEFAX: 202-371-2540 <br><br> (2) INFORMATION FOR SEQ ID NO:l: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 1442 base pairs <br><br> -53- <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double &lt;D) TOPOLOGY: both <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (ix) FEATURE: <br><br> (A) NAME/KEY: CDS <br><br> (B) LOCATION: 15..1139 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1: <br><br> TTTTTTTTTT TGAG ATG GAG TTT TCG CTC TTG TTG CCC AGG CTG GAG TGC 50 <br><br> Met Glu Phe Ser Leu Leu Leu Pro Arg Leu Glu Cys 1 5 10 <br><br> AAT GGC GCA ATC TCA GCT CAC CGC AAC CTC CGC CTC CCG GGT TCA AGC 98 <br><br> Asn Gly Ala lie Ser Ala His Arg Asn Leu Arg Leu Pro Gly Ser Ser 15 20 25 <br><br> GAT TCT CCT GCC TCA GCC TCC CCA GTA GCT GGG ATT ACA GGC ATG TGC 14 6 <br><br> Asp Ser Pro Ala Ser Ala Ser Pro Val Ala Gly lie Thr Gly Met Cys 30 35 40 <br><br> ACC CAC GCT CGG CTA ATT TTG TAT TTT TTT TTA GTA GAG ATG GAG TTT 194 <br><br> Thr His Ala Arg Leu lie Leu Tyr Phe Phe Leu Val Glu Met Glu Phe 45 50 55 60 <br><br> CTC CAT GTT GGT CAG GCT GGT CTC GAA CTC CCG ACC TCA GAT GAT CCC 24 2 <br><br> Leu His Val Gly Gin Ala Gly Leu Glu Leu Pro Thr Ser Asp Asp Pro 65 70 75 <br><br> TCC GTC TCG GCC TCC CAA AGT GCT AGA TAC AGG ACT GGC CAC CAT GCC 290 <br><br> Ser Val Ser Ala Ser Gin Ser Ala Arg Tyr Arg Thr Gly His His Ala 80 85 90 <br><br> CGG CTC TGC CTG GCT AAT TTT TGT GGT AGA AAC AGG GTT TCA CTG ATG 338 <br><br> Arg Leu Cys Leu Ala Asn Phe Cys Gly Arg Asn Arg Val Ser Leu Met 95 100 105 <br><br> TGC CCA AGC TGG TCT CCT GAG CTC AAG CAG TCC ACC TGC CTC AGC CTC 38 6 <br><br> Cys Pro Ser Trp Ser Pro Glu Leu Lys Gin Ser Thr Cys Leu Ser Leu 110 115 120 <br><br> CCA AAG TGC TGG GAT TAC AGG CGT GCA GCC GTG CCT GGC CTT TTT ATT 4 34 <br><br> Pro Lys Cys Trp Asp Tyr Arg Arg Ala Ala Val Pro Gly Leu Phe lie 125 130 135 140 <br><br> TTA TTT TTT TTA AGA CAC AGG TGT CCC ACT CTT ACC CAG GAT GAA GTG 4 82 <br><br> Leu Phe Phe Leu Arg His Arg Cys Pro Thr Leu Thr Gin Asp Glu Val <br><br> 145 150 155 _ <br><br> CAG TGG TGT GAT CAC AGC TCA CTG CAG CCT TCA ACT CCT GAG ATC AAG 530 <br><br> Gin Trp Cys Asp His Ser Ser Leu Gin Pro Ser Thr Pro Glu lie Lys <br><br> -54- <br><br> 160 <br><br> 165 <br><br> 170 <br><br> CAT CCT CCT GCC TCA GCC TCC CAA GTA GCT GGG ACC AAA GAC ATG CAC His Pro Pro Ala Ser Ala Ser Gin Val Ala Gly Thr Lys Asp Met His 175 180 185 <br><br> 578 <br><br> CAC TAC ACC TGG CTA ATT TTT ATT TTT ATT TTT AAT TTT TTG AGA CAG His Tyr Thr Trp Leu lie Phe lie Phe lie Phe Asn Phe Leu Arg Gin 190 195 200 <br><br> 626 <br><br> AGT CTC AAC TCT GTC ACC CAG GCT GGA GTG CAG TGG CGC AAT CTT GGC 674 <br><br> Ser Leu Asn Ser Val Thr Gin Ala Gly Val Gin Trp Arg Asn Leu Gly 205 210 215 220 <br><br> TCA CTG CAA CCT CTG CCT CCC GGG TTC AAG TTA TTC TCC TGC CCC AGC 722 <br><br> Ser Leu Gin Pro Leu Pro Pro Gly Phe Lys Leu Phe Ser Cys Pro Ser 225 230 235 <br><br> CTC CTG AGT AGC TGG GAC TAC AGG CGC CCA CCA CGC CTA GCT AAT TTT 770 <br><br> Leu Leu Ser Ser Trp Asp Tyr Arg Arg Pro Pro Arg Leu Ala Asn Phe 240 245 250 <br><br> TTT GTA TTT TTA GTA GAG ATG GGG TTC ACC ATG TTC GCC AGG TTG ATC 818 <br><br> Phe Val Phe Leu Val Glu Met Gly Phe Thr Met Phe Ala Arg Leu lie 255 260 265 <br><br> TTG ATC TCT GGA CCT TGT GAT CTG CCT GCC TCG GCC TCC CAA AGT GCT 8 66 <br><br> Leu lie Ser Gly Pro Cys Asp Leu Pro Ala Ser Ala Ser Gin Ser Ala 270 275 280 <br><br> GGG ATT ACA GGC GTG AGC CAC CAC GCC CGG CTT ATT TTT AAT TTT TGT 914 <br><br> Gly lie Thr Gly Val Ser His His Ala Arg Leu lie Phe Asn Phe Cys 285 290 295 300 <br><br> TTG TTT GAA ATG GAA TCT CAC TCT GTT ACC CAG GCT GGA GTG CAA TGG 962 <br><br> Leu Phe Glu Met Glu Ser His Ser Val Thr Gin Ala Gly Val Gin Trp 305 310 315 <br><br> CCA AAT CTC GGC TCA CTG CAA CCT CTG CCT CCC GGG CTC AAG CGA TTC 1010 <br><br> Pro Asn Leu Gly Ser Leu Gin Pro Leu Pro Pro Gly Leu Lys Arg Phe 320 325 330 <br><br> TCC TGT CTC AGC CTC CCA AGC AGC TGG GAT TAC GGG CAC CTG CCA CCA 1058 <br><br> Ser Cys Leu Ser Leu Pro Ser Ser Trp Asp Tyr Gly His Leu Pro Pro 335 340 345 <br><br> CAC CCC GCT AAT TTT TGT ATT TTC ATT AGA GGC GGG GTT TCA CCA TAT 1106 <br><br> His Pro Ala Asn Phe Cys lie Phe lie Arg Gly Gly Val Ser Pro Tyr 350 355 360 <br><br> TTG TCA GGC TGG TCT CAA ACT CCT GAC CTC AGG TGACCCACCT GCCTCAGCCT 115 9 Leu Ser Gly Trp Ser Gin Thr Pro Asp Leu Arg 365 370 375 <br><br> TCCAAAGTGC TGGGATTACA GGCGTGAGCC ACCTCACCCA GCCGGCTAAT TTAGATAAAA 1219 <br><br> -55- <br><br> AAATATGTAG CAATGGGGGG TCTTGCTATG TTGCCCAGGC TGGTCTCAAA CTTCTGGCTT 127 9 <br><br> CATGCAATCC TTCCAAATGA GCCACAACAC CCAGCCAGTC ACATTTTTTA AACAGTTACA 1339 <br><br> TCTTTATTTT AGTATACTAG AAAGTAATAC AATAAACATG TCAAACCTGC AAATTCAGTA 1399 <br><br> GTAACAGAGT TCTTTTATAA CTTTTAAACA AAGCTTTAGA GCA 144 2 <br><br> (2) INFORMATION FOR SEQ ID NO: 2: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 37 5 amino acids <br><br> (B) TYPE: amino acid (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: protein <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: <br><br> Met Glu Phe Ser Leu Leu Leu Pro Arg Leu Glu Cys Asn Gly Ala lie 1 5 10 15 <br><br> Ser Ala His Arg Asn Leu Arg Leu Pro Gly Ser Ser Asp Ser Pro Ala 20 25 30 <br><br> Ser Ala Ser Pro Val Ala Gly lie Thr Gly Met Cys Thr His Ala Arg 35 40 45 <br><br> Leu lie Leu Tyr Phe Phe Leu Val Glu Met Glu Phe Leu His Val Gly 50 55 60 <br><br> Gin Ala Gly Leu Glu Leu Pro Thr Ser Asp Asp Pro Ser Val Ser Ala 65 70 75 80 <br><br> Ser Gin Ser Ala Arg Tyr Arg Thr Gly His His Ala Arg Leu Cys Leu 85 90 95 <br><br> Ala Asn Phe Cys Gly Arg Asn Arg Val Ser Leu Met Cys Pro Ser Trp 100 105 110 <br><br> Ser Pro Glu Leu Lys Gin Ser Thr Cys Leu Ser Leu Pro Lys Cys Trp 115 120 125 <br><br> Asp Tyr Arg Arg Ala Ala Val Pro Gly Leu Phe lie Leu Phe Phe Leu 130 135 140 <br><br> Arg His Arg Cys Pro Thr Leu Thr Gin Asp Glu Val Gin Trp Cys Asp 145 150 155 160 <br><br> His Ser Ser Leu Gin Pro Ser Thr Pro Glu lie Lys His Pro Pro Ala 165 170 175 <br><br> Ser Ala Ser Gin Val Ala Gly Thr Lys Asp Met His His Tyr Thr Trp 180 "" 185 190 <br><br> -56- <br><br> Leu lie Phe lie Phe lie Phe Asn Phe Leu Arg Gin Ser Leu Asn Ser 195 200 205 <br><br> Val Thr Gin Ala Gly Val Gin Trp Arg Asn Leu Gly Ser Leu Gin Pro 210 215 220 <br><br> Leu Pro Pro Gly Phe Lys Leu Phe Ser Cys Pro Ser Leu Leu Ser Ser 225 230 235 240 <br><br> Trp Asp Tyr Arg Arg Pro Pro Arg Leu Ala Asn Phe Phe Val Phe Leu 245 250 255 <br><br> Val Glu Met Gly Phe Thr Met Phe Ala Arg Leu lie Leu lie Ser Gly 260 265 270 <br><br> Pro Cys Asp Leu Pro Ala Ser Ala Ser Gin Ser Ala Gly lie Thr Gly 275 280 285 <br><br> Val Ser His His Ala Arg Leu lie Phe Asn Phe Cys Leu Phe Glu Met 290 295 300 <br><br> Glu Ser His Ser Val Thr Gin Ala Gly Val Gin Trp Pro Asn Leu Gly 305 310 315 320 <br><br> Ser Leu Gin Pro Leu Pro Pro Gly Leu Lys Arg Phe Ser Cys Leu Ser 325 330 335 <br><br> Leu Pro Ser Ser Trp Asp Tyr Gly His Leu Pro Pro His Pro Ala Asn 340 345 350 <br><br> Phe Cys lie Phe lie Arg Gly Gly Val Ser Pro Tyr Leu Ser Gly Trp 355 360 365 <br><br> Ser Gin Thr Pro Asp Leu Arg 370 375 <br><br> (2) INFORMATION FOR SEQ ID NO:3: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 1381 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: double <br><br> (D) TOPOLOGY: both <br><br> (ii) MOLECULE TYPE: cDNA <br><br> 60 120 180 <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: <br><br> TTTTTTTTTT GAGATGGAGT TTTCGCTCTT GTTGCCCAGG CTGGAGTGCA ATGGCGCAAT CTCAGCTCAC CGCAACCTCC GCCTCCCGGG TTCAAGCGAT TCTCCTGCCT CAGCCTCCCC AGTAGCTGGG ATTACAGGCA TGTGCACCAC GCTCGGCTAA TTTTGTATTT TTTTTTAGTA <br><br> -57- <br><br> GAGATGGAGT TTAACTCCAT GTTGGTCAGG CTGGTCTCGA ACTCCCGACC T CAGAT GAT C 24 0 <br><br> TCCCGTCTCG GCCTGCCCAA AGTGCTGAGA TTACAGGCAT GAGCCACCAT GCCCGGCCTC 300 <br><br> TGCCTGGCTA ATTTTTGTGG TAGAAACAGG GTTTCACTGA TGTTGCCCAA GCTGGTCTCC 360 <br><br> TGAGCTCAAG CAGTCCACCT GCCTCAGCCT CCCAAAGTGC TGGGATTACA GGCGTCAGCC 4 20 <br><br> GTGCCTGGCC TTTTTATTTT ATTTTTTTTA AGACACAGGT GTACCACTCT TACCCAGGAT 4 80 <br><br> GAAGTGCAGT GGTGTGATCA CAGCTCACTG CAGCCTTCAA CTCCTGAGAT CAAGCAATCC 54 0 <br><br> TCCTGCCTCA GCCTCCCAAG TAGCTGGGAC CAAAGACATG CACCACTACA CCTGGTAATT 600 <br><br> TTTATTTTTA TTTTTAATTT TTTGAGACAG AGTCTCACTC TGTCACCCAG GCTGGAGTGC 660 <br><br> AGTGGCGCAA TCTTGGCTCA CTGCAACCTC TGCCTCCCGG GTTCAAGTTA TTCTCCTGCC 7 20 <br><br> CCAGCCTCCT GAGTAGCTGG GACTACAGGC GCCCACCACG CCTAGCTAAT TTTTTTGTAT 780 <br><br> TTTTAGTAGA GATGGGGTTT CACCATGTTC GCCAGGTTGA TCTTGATCTC TTGACCTTGT 84 0 <br><br> GATCTGCCTG CCTCGGCCTA CCCAAAGTGC TGGGATTACA GGTCGTGACT CCACGCCGGC 900 <br><br> CTATTTTTAA TTTTTGTTTG TTTGAAATGG AATCTCACTC TGTTACCCAG GTCGGAGTGC 960 <br><br> AATGGCAAAT CTCGGCTACT CGCAACCTCT GCCTCCCGGG TCAAGCGATT CTCCTGTCTC 1020 <br><br> AGCCTCCCAA GCAGCTGGGA TTACGGGACC TGCACCACAC CCCGCTAATT TTTGTATTTT 1080 <br><br> CATTAGAGGC GGGTTTACCA TATTTGTCAG GCTGGGTCTC AAACTCCTGA CCTCAGGTGA 114 0 <br><br> CCCACCTGCC TCAGCCTTCC AAAGTGCTGG GATTACAGGC GTGAGCCACC TCACCCAGCC 1200 <br><br> GGCTAATTTG GAATAAAAAA TATGTAGCAA TGGGGGTCTG CTATGTTGCC CAGGCTGGTC 12 60 <br><br> TCAAACTTCT GGCTTCAGTC AATCCTTCCA AATGAGCCAC AACACCCAGC CAGTCACATT 1320 <br><br> TTTTAAACAG TTACATCTTT ATTTTAGTAT ACTAGAAAGT AATACAATAA ACATGTCAAA 1380 <br><br> C 1381 (2) INFORMATION FOR SEQ ID NO:4: <br><br> (i) SEQUENCE CHARACTERISTICS : <br><br> (A) LENGTH: 1418 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: both <br><br> (D) TOPOLOGY: both <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: <br><br> -58- <br><br> tttttttttt gagatggagt TTTCGCTCTT GTTGCCCAGG CTGGAGTGCA ATGGCGCAAT 60 <br><br> CTCAGCTCAC CGCAACCTCC GCCTCCCGGG TTCAAGCGAT TCTCCTGCCT CAGCCTCCCC 120 <br><br> AGTAGGCTGG GATTACAGGC ATGTGCACCA CGCTCGGCTA ATTTTGTATT TTTTTTTAGT 180 <br><br> AGAGATGGAG TTTCTCCATG TTGGTCAGGC TGGTCTCGAA CTCCGACCTC AGATGATCCT • 24 0 <br><br> CCCGTCTCGG CCTCCCAAAG TGCTAGATAC AGGACTGAGC ACCATGCCCG GCCTCTGCCT 300 <br><br> GGCTAATTTT TGTGGTAGAA ACAGGGTTTC ACTGATGTGC CCAAGCTGGT CTCCTGAGCT 360 <br><br> CAAGCAGTCC ACCTGCCTCA GCCTCCCAAA GTGCTGGGAT TACAGGCGTG CAGCCGTGCC 4 20 <br><br> TGGCCTTTTT ATTTTATTTT TTTTAAGACA CAGGTGTCCC ACTCTTACCC AGGATGAAGT 4 80 <br><br> GCAGTGGTGT GATCACAGCT CACTGCAGCC TTCAACTCTG AGATCAAGCA TCCTCCTGCC 540 <br><br> TCAGCCTCCC AAAGTAGCTG GGACCAAAGA CATGCACCAC TACACCTGGC TAATTTTTAT 600 <br><br> TTTTATTTTT AATTTTTTGA GACAGAGTCT CAACTCTGTC ACCCAGGCTG GAGTGCAGTG 660 <br><br> GCGCAATCTT GGCTCACTGC AACCTCTGCC TCCCGGGTTC AAGTTATTCT CCTGCCCCAG 7 20 <br><br> CCTCCTGAGT AGCTGGGACT ACAGGCGCCC ACCACGCCTA GCTAATTTTT TTGTATTTTT 780 <br><br> AG TAGAGATG GGGTTTCACC ATGTTCGCCA GGTTGATGCT AGATCTCTTG ACCTTGTGAT 8 40 <br><br> CTGCCTGCCT CGGCCTCCCA AAGTGCTGGG ATTACAGGAC GTGACGCCCA CCGCCCGGCC 900 <br><br> TATTTTTAAT TTTTGTTTGT TTGAAATGGA ATCTCACTCT GTTACCCAGG CTGGAGTGCA 960 <br><br> ATGGCCAAAT CTCGGCTCAC TGCAACCTCT GCCTCCCGGG CTCAAGCGAT TCTCCTGTCT 1020 <br><br> CAGCCTCCCA AGCAGCTGGG ATTACGGGCA CCTGCACCAC ACCCCGCTAA TTTTTGTATT 1080 <br><br> TTCATTAGAG GCGGGGTTTC ACCATATTTG TCAGGCTGGT CTCAAACTCC TGACCTCAGG 114 0 <br><br> TGACCCACCT GCCTCAGCCT TCCAAAGTGC TGGGATTACA GGCGTGACGC CTCACCCAGC 1200 <br><br> CGGCTAATTT AGATAAAAAA ATATGTAGCA ATGGGGGGTC TTGCTATGTT GCCCAGGCTG 1260 <br><br> GTCTCAAACT TCTGGCTTCA TGCAATCCTT CCAAATGAGC CACAACACCC AGCCAGTCAC 1320 <br><br> ATTTTTAAAC AGTTACATCT TTATTTTAGT ATACTAGAAA GTGATACGAT AACATGGCGG 1380 <br><br> AACCTGCAAA TTCGAGTAGT ACAGAGTCTT TTATAACT 1418 (2) INFORMATION FOR SEQ ID NO:5: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 22 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> -59- <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5: <br><br> TGTCCCACTC TTACCCAGGA TG 22 <br><br> (2) INFORMATION FOR SEQ ID NO:6: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 24 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6: <br><br> AAGCAGGCAG ATCACAAGGT CCAG 24 <br><br> (2! INFORMATION FOR SEQ ID NO:7: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 20 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: <br><br> AATGGATGAC GATATCGCTG 20 <br><br> (2) INFORMATION FOR SEQ ID NO:8: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 19 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> -60- <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8 ATGAGGTAGT CTGTCAGGT (2) INFORMATION FOR SEQ ID NO: 9: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 30 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: TTCATCCTGG GTAAGAGTGG GACACCTGTG (2) INFORMATION FOR SEQ ID NO:10: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 26 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:10 TGGTGCATGT CTTTGGTCCC AGCTAC (2) INFORMATION FOR SEQ ID NO:11: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 30 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11 ATCAACCTGG CGAACATGGT GAACCCCATC (2) INFORMATION FOR SEQ ID NO:12: <br><br> -61- <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 14 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: CACTGCACTT NCCA <br><br> (2) INFORMATION FOR SEQ ID NO:13: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 14 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: CCAGGTGTAG NCCA <br><br> (2) INFORMATION FOR SEQ ID NO:14: <br><br> (i) SEQUENCE CHARACTERISTICS: <br><br> (A) LENGTH: 14 base pairs <br><br> (B) TYPE: nucleic acid <br><br> (C) STRANDEDNESS: single <br><br> (D) TOPOLOGY: linear <br><br> (ii) MOLECULE TYPE: cDNA <br><br> (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: CAAGGTCCAG NCCA <br><br> -61,1- <br><br> Applicant's or agent's file <br><br> International applicBKijiiD^y '" -- <br><br> 98.-'a36S5 <br><br> reference number 0609.437PC01 <br><br> TBA <br><br> INDICATIONS RELATING TO A DEPOSITED MICROORGANISM (PCT Rule 13tos) <br><br> A. The indications made below relate to the microorganism refen-ed to in the description on page 5 , line 11 . <br><br> B. IDENTIFICATION OF DEPOSIT <br><br> Funher deposits are identified on an additional sheet Q <br><br> Name of depositary institution <br><br> AMERICAN TYPE CULTURE COLLECTION <br><br> Address of depositary institution (including postal code and country) <br><br> 12301 Parklawn Drive <br><br> Rockville, Maryland 20852 <br><br> United States of America <br><br> Date of deposit <br><br> Accession Number <br><br> March 16, 1993 <br><br> 69262 <br><br> C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information is continued on an additional sheet D <br><br> Escherichia coii: AD10-7-DH1 <br><br> • <br><br> D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (if the indications are no! for all designated States) <br><br> E. SEPARATE FURNISHING OF INDICATIONS (leave blank if not applicable) <br><br> The indications listed below wiil be submitted to the intemationai Bureau later (specify the general nature of the indications, e.g., <br><br> "Accession Number of Deposit") <br><br> For receiving Office use only <br><br> □ This sheet was received with the intemationai application <br><br> Authorized officer <br><br> For Intemationai Bureau use only <br><br> Q This sheet was received by the International Bureau on: <br><br> Authorized officer <br><br> Form PCT/RO/I34 (July 1992) <br><br> 437dep.jp <br><br></p> </div>

Claims (5)

<div class="application article clearfix printTableText" id="claims"> <p lang="en"> -62-<br><br> WHAT WE CLAIM IS:<br><br>
1. A method to treat or prevent dementias of the Alzheimer's type of neuronal degeneration; or to treat or prevent neuroectodermal tumors, malignant astrocytomas, or glioblastomas, comprising administering to a non-human animal in need thereof an antisense oligonucleotide, a ribozyme or a triple helix-forming oligonucleotide selected from<br><br> (a) an antisense oligonucleotide which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l;<br><br> (b) a ribozyme comprising a target sequence which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ID No.l;<br><br> (c) an oligodeoxynucleotide that forms triple stranded regions with the a region of AD7c-NTP coding nucleic acid and having the sequence 3'X5'-L-5'X3', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq. ID No.l, and wherein L represents an oligonucleotide linker or a bond; and<br><br> (d) an oligodeoxynucleotide that forms triple stranded regions with a region of AD7c-NTP coding nucleic acid and having the sequence 5'X3'-L-3'X5', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq ED No. 1, and wherein L represents an oligonucleotide linker or a bond.<br><br>
2. The method of claim 1, wherein said antisense oligonucleotide, ribozyme or triple-helix forming oligonucleotide is administered to said animal with a pharmaceutically acceptable carrier as part of a pharmaceutical composition.<br><br>
3- A method as defined in claim 1 substantially as herein described with reference to any example thereof.<br><br> intellectual property office of n.z<br><br> 2 6 AUG 20(ft RECEIVED<br><br> -63-<br><br>
4. A use, in the preparation of a medicament for treating or preventing a condition selected from the group comprising dementias of the Alzheimer's type of neuronal degeneration, neuroectodermal tumors, malignant astrocytomas, and glioblastomas in a patient in need thereof, of an antisense oligonucleotide, a ribozyme or a triple helix-forming oligonucleotide selected from<br><br> (a) an antisense oligonucleotide which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ED No.l;<br><br> (b) a ribozyme comprising a target sequence which is complementary to an NTP mRNA sequence corresponding to nucleotides 150-1139 of Seq. ED No.l;<br><br> (c) an oligodeoxynucleotide that forms triple stranded regions with the a region of AD7c-NTP coding nucleic acid and having the sequence 3'X5'-L-5'X3', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq. ID No.l, and wherein L represents an oligonucleotide linker or a bond; and<br><br> (d) an oligodeoxynucleotide that forms triple stranded regions with a region of AD7c-NTP coding nucleic acid and having the sequence 5'X3'-L-3'X5', wherein X comprises an AD7c-NTP nucleic acid sequence corresponding to nucleotides 150-1139 of Seq ED No.l, and wherein L represents an oligonucleotide linker or a bond.<br><br>
5. A use as claimed in claim 4, substantially as herein described with reference to any example thereof.<br><br> END OF CLAIMS<br><br> intellectual property office of n.z<br><br> 2 6 AUG 200*1<br><br> </p> </div>
NZ508018A 1997-02-26 2000-11-08 DNA construct containing AD7c-NTP sequence controlled by neurospecific promoter to treat Alzheimer's disease and neural tumours NZ508018A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US3890897P 1997-02-26 1997-02-26

Publications (1)

Publication Number Publication Date
NZ508018A true NZ508018A (en) 2004-10-29

Family

ID=21902584

Family Applications (2)

Application Number Title Priority Date Filing Date
NZ337445A NZ337445A (en) 1997-02-26 1998-02-26 Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of alzheimer's disease
NZ508018A NZ508018A (en) 1997-02-26 2000-11-08 DNA construct containing AD7c-NTP sequence controlled by neurospecific promoter to treat Alzheimer's disease and neural tumours

Family Applications Before (1)

Application Number Title Priority Date Filing Date
NZ337445A NZ337445A (en) 1997-02-26 1998-02-26 Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of alzheimer's disease

Country Status (8)

Country Link
US (3) US7138380B2 (en)
EP (1) EP0975651A4 (en)
JP (1) JP4194664B2 (en)
KR (1) KR20000075748A (en)
AU (1) AU749348B2 (en)
CA (1) CA2282729A1 (en)
NZ (2) NZ337445A (en)
WO (1) WO1998038204A1 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6271360B1 (en) * 1999-08-27 2001-08-07 Valigen (Us), Inc. Single-stranded oligodeoxynucleotide mutational vectors
CN1635906A (en) * 2001-05-04 2005-07-06 尼莫克斯股份有限公司 Method of preventing cell death using antibodies to neural thread proteins
CA2448348C (en) 2001-05-25 2013-07-16 Nymox Corporation Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
US6770797B2 (en) 2001-06-01 2004-08-03 Rhode Island Hospital Non-Transgenic nonhuman model for Alzheimer's Disease using a AD7c-NTP nucleic acid
KR101005130B1 (en) 2001-07-19 2011-01-04 니목스 코포레이션 Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
EP1847550A3 (en) * 2001-07-19 2008-01-09 Nymox Corporation Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
DE10154399A1 (en) * 2001-11-06 2003-05-15 Basf Lynx Bioscience Ag Process for the identification of active substances for the modulation of pip92-mediated apoptosis
US7317077B2 (en) 2001-11-16 2008-01-08 Nymox Pharmaceutical Corporation Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
EP1714979A3 (en) * 2001-11-16 2007-04-25 Nymox Corporation Peptides effective in the treatment of tumors and other conditions requiring the removal or destruction of cells
GB0419124D0 (en) * 2004-08-27 2004-09-29 Proteome Sciences Plc Methods and compositions relating to Alzheimer's disease
US7544771B2 (en) 2005-02-23 2009-06-09 Nymox Corporation Protein and its use in diagnosing Alzheimer's disease
US7768422B2 (en) * 2006-09-06 2010-08-03 Carmen Jr Lawrence R Method of restoring a remote wireless control device to a known state
EP3029061B1 (en) 2008-06-18 2017-09-13 The Texas A&M University System Mesenchymal stem cells, compositions, and methods for treatment of cardiac tissue damage
EP3044326B1 (en) * 2013-09-13 2017-12-06 F. Hoffmann-La Roche AG Application of oligo-dt molecules to avoid generation of high molecular pcr products induced by polya-carrier
US11628202B2 (en) 2015-07-24 2023-04-18 Nymox Corporation Methods of reducing the need for surgery in patients suffering from benign prostatic hyperplasia
US10183058B2 (en) 2016-06-17 2019-01-22 Nymox Corporation Method of preventing or reducing the progression of prostate cancer
US10172910B2 (en) 2016-07-28 2019-01-08 Nymox Corporation Method of preventing or reducing the incidence of acute urinary retention
US10532081B2 (en) 2016-09-07 2020-01-14 Nymox Corporation Method of ameliorating or preventing the worsening or the progression of symptoms of BPH
US10335453B2 (en) 2017-03-01 2019-07-02 Nymox Corporation Compositions and methods for improving sexual function

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
FR2604438B1 (en) 1986-09-26 1988-12-23 Centre Nat Rech Scient NOVEL COUPLING CONJUGATES BETWEEN RNA OR DNA SEQUENCES AND A PROTEIN, THEIR PREPARATION PROCESS AND THEIR BIOLOGICAL APPLICATION
EP0383803B1 (en) 1987-10-28 2000-05-03 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates
US5948634A (en) 1988-12-21 1999-09-07 The General Hospital Coporation Neural thread protein gene expression and detection of alzheimer's disease
JP3309972B2 (en) 1988-12-21 2002-07-29 ザ・ジェネラル・ホスピタル・コーポレイション Methods for detecting nervous system disorders or dysfunction
US5168053A (en) 1989-03-24 1992-12-01 Yale University Cleavage of targeted RNA by RNAase P
DE69123979T2 (en) 1990-10-12 1997-04-30 Max Planck Gesellschaft MODIFIED RIBOZYMS
DK0697893T3 (en) * 1993-04-20 2006-02-20 Gen Hospital Corp Nerve Thread Protein Gene Expression and Detection of Alzheimer's Disease
WO1996040895A1 (en) 1995-06-07 1996-12-19 Athena Neurosciences, Inc. Method for identifying alzheimer's disease therapeutics using transgenic animal models
US5648888A (en) * 1995-12-27 1997-07-15 Hydro-Quebec Power distribution substation
ATE414149T1 (en) 1996-11-20 2008-11-15 Univ Yale SURVIVIN, A PROTEIN THAT INHIBITS CELLULAR APOPTOSIS AND ITS MODULATION

Also Published As

Publication number Publication date
EP0975651A4 (en) 2005-03-09
US7291454B2 (en) 2007-11-06
AU6667598A (en) 1998-09-18
US20020104108A1 (en) 2002-08-01
US7045616B2 (en) 2006-05-16
US20020129391A1 (en) 2002-09-12
US20030066097A1 (en) 2003-04-03
AU749348B2 (en) 2002-06-27
JP2001513777A (en) 2001-09-04
NZ337445A (en) 2000-12-22
CA2282729A1 (en) 1998-09-03
KR20000075748A (en) 2000-12-26
JP4194664B2 (en) 2008-12-10
EP0975651A1 (en) 2000-02-02
US7138380B2 (en) 2006-11-21
WO1998038204A1 (en) 1998-09-03

Similar Documents

Publication Publication Date Title
AU749348B2 (en) Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of Alzheimer&#39;s disease
WO1998038204A9 (en) Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of alzheimer&#39;s disease
Lim et al. FTDP-17 mutations in tau transgenic mice provoke lysosomal abnormalities and Tau filaments in forebrain
Von Koch et al. Generation of APLP2 KO mice and early postnatal lethality in APLP2/APP double KO mice
US7393994B2 (en) Transgenic mouse model for neurodegenerative diseases
EP2220251B1 (en) Method for the diagnosis of pathologies characterised by the anomalous deposition of amyloid in organs and tissues by detecting a mutation at position 673 in app770, and vector and peptide for use in the therapy of said pathologies
US7309812B2 (en) Perlecan transgenic animals and methods of identifying compounds for the treatment of amyloidoses
CA2198451A1 (en) Transgenic animal expressing a familial form of human amyloid precursor protein
JP2004261187A (en) Alzheimer&#39;s disease model
Tanaka et al. Murine model of Alexander disease: analysis of GFAP aggregate formation and its pathological significance
JP5070236B2 (en) Transgenic animal model of neurodegenerative disorder
US7226730B1 (en) Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of Alzheimer&#39;s Disease
US20030033621A1 (en) Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of Alzheimer&#39;s disease
AU2002312033B2 (en) Inhibition of neurodegeneration
EP1670943B1 (en) Diagnostic and therapeutic use of a sulfotransferase for alzheimer&#39;s disease
AU2002312033A1 (en) Inhibition of neurodegeneration
EP2207885B1 (en) Quadruple transgenic non-human animal
CA2458193A1 (en) Development of transgenic model for interventions in neurodegenerative diseases
WO1994024266A1 (en) Transgenic animal models for alzheimer&#39;s disease
JP2007514406A (en) Diagnostic and therapeutic uses of human DAX-1 gene and protein for neurodegenerative diseases
Beech On the causes of Alzheimer's disease: Investigations using transgenic mouse model systems

Legal Events

Date Code Title Description
RENW Renewal (renewal fees accepted)
PSEA Patent sealed
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
EXPY Patent expired